1
|
Zhao Y, Wang X, Yang X, Li J, Han B. Insights into the history and trends of nanotechnology for the treatment of hepatocellular carcinoma: a bibliometric-based visual analysis. Discov Oncol 2025; 16:484. [PMID: 40192866 PMCID: PMC11977073 DOI: 10.1007/s12672-025-02145-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/13/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Nanotechnology has great potential and advantages in the treatment of hepatocellular carcinoma (HCC), but the research trends and future directions are not yet clear. OBJECTIVES Analyze the development trajectory, research hotspots, and future trends of nanotechnology and HCC research globally in the past 20 years, providing a more comprehensive and intuitive reference for researchers in this field. METHODS Retrieve relevant literature on nanotechnology and HCC research in the Web of Science (WOS) Core Collection database, and conduct bibliometric analysis using software such as CiteSpace, VOSviewer, and SCImago Graphica. RESULTS A total of 852 English publications meeting the criteria were retrieved from the WOS database, with an overall increasing trend in the number of publications and citation frequency over the years. China leads in the number of publications and international collaborations, followed by the USA and India. The most influential research institution is the Chinese Academy of Sciences, the most influential scholar/team is the Rahman, Mahfoozur team, and the journal with the most publications is the International Journal of Nanomedicine. A comprehensive analysis reveals that the current main research directions include new types of nanoparticles, targeted drug delivery systems, photothermal/photodynamic therapy, gene delivery systems, diagnostics, and imaging. It is anticipated that further collaboration among scholars, institutions, and countries will accelerate the development of nanotechnology in the field of HCC research. CONCLUSION This study provides an in-depth analysis of the research status and development trends of nanotechnology in treating HCC from a bibliometric perspective, offering possible guidance for researchers to explore hot topics and frontiers, select suitable journals, and partners in this field.
Collapse
Affiliation(s)
- Yulei Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Xingxin Wang
- College of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaoman Yang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Jiaheng Li
- College of Health, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Bingbing Han
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
2
|
Sailo BL, Chauhan S, Hegde M, Girisa S, Alqahtani MS, Abbas M, Goel A, Sethi G, Kunnumakkara AB. Therapeutic potential of tocotrienols as chemosensitizers in cancer therapy. Phytother Res 2025; 39:1694-1720. [PMID: 38353331 DOI: 10.1002/ptr.8131] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/29/2023] [Accepted: 01/15/2024] [Indexed: 04/23/2025]
Abstract
Chemoresistance is the adaptation of cancer cells against therapeutic agents. When exhibited by cancer cells, chemoresistance helps them to avoid apoptosis, cause relapse, and metastasize, making it challenging for chemotherapeutic agents to treat cancer. Various strategies like dosage modification of drugs, nanoparticle-based delivery of chemotherapeutics, antibody-drug conjugates, and so on are being used to target and reverse chemoresistance, one among such is combination therapy. It uses the combination of two or more therapeutic agents to reverse multidrug resistance and improve the effects of chemotherapy. Phytochemicals are known to exhibit chemosensitizing properties and are found to be effective against various cancers. Tocotrienols (T3) and tocopherols (T) are natural bioactive analogs of vitamin E, which exhibit important medicinal value and potential curative properties apart from serving as an antioxidant and nutrient supplement. Notably, T3 exhibits a variety of pharmacological activities like anticancer, anti-inflammatory, antiproliferative, and so on. The chemosensitizing property of tocotrienol is exhibited by modulating several signaling pathways and molecular targets involved in cancer cell survival, proliferation, invasion, migration, and metastasis like NF-κB, STATs, Akt/mTOR, Bax/Bcl-2, Wnt/β-catenin, and many more. T3 sensitizes cancer cells to chemotherapeutic drugs including cisplatin, doxorubicin, and paclitaxel increasing drug concentration and cytotoxicity. Discussed herewith are the chemosensitizing properties of tocotrienols on various cancer cell types when combined with various drugs and biological molecules.
Collapse
Affiliation(s)
- Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Suravi Chauhan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Arul Goel
- University of California Santa Barbara, Santa Barbara, California, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
3
|
Bano N, Parveen S, Saeed M, Siddiqui S, Abohassan M, Mir SS. Drug Repurposing of Selected Antibiotics: An Emerging Approach in Cancer Drug Discovery. ACS OMEGA 2024; 9:26762-26779. [PMID: 38947816 PMCID: PMC11209889 DOI: 10.1021/acsomega.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/19/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024]
Abstract
Drug repurposing is a method of investigating new therapeutic applications for previously approved medications. This repurposing approach to "old" medications is now highly efficient, simple to arrange, and cost-effective and poses little risk of failure in treating a variety of disorders, including cancer. Drug repurposing for cancer therapy is currently a key topic of study. It is a way of exploring recent therapeutic applications for already-existing drugs. Theoretically, the repurposing strategy has various advantages over the recognized challenges of creating new molecular entities, including being faster, safer, easier, and less expensive. In the real world, several medications have been repurposed, including aspirin, metformin, and chloroquine. However, doctors and scientists address numerous challenges when repurposing drugs, such as the fact that most drugs are not cost-effective and are resistant to bacteria. So the goal of this review is to gather information regarding repurposing pharmaceuticals to make them more cost-effective and harder for bacteria to resist. Cancer patients are more susceptible to bacterial infections. Due to their weak immune systems, antibiotics help protect them from a variety of infectious diseases. Although antibiotics are not immune boosters, they do benefit the defense system by killing bacteria and slowing the growth of cancer cells. Their use also increases the therapeutic efficacy and helps avoid recurrence. Of late, antibiotics have been repurposed as potent anticancer agents because of the evolutionary relationship between the prokaryotic genome and mitochondrial DNA of eukaryotes. Anticancer antibiotics that prevent cancer cells from growing by interfering with their DNA and blocking growth of promoters, which include anthracyclines, daunorubicin, epirubicin, mitoxantrone, doxorubicin, and idarubicin, are another type of FDA-approved antibiotics used to treat cancer. According to the endosymbiotic hypothesis, prokaryotes and eukaryotes are thought to have an evolutionary relationship. Hence, in this study, we are trying to explore antibiotics that are necessary for treating diseases, including cancer, helping people reduce deaths associated with various infections, and substantially extending people's life expectancy and quality of life.
Collapse
Affiliation(s)
- Nilofer Bano
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow 226026, India
| | - Sana Parveen
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow 226026, India
| | - Mohd Saeed
- Department
of Biology, College of Sciences, University
of Hail, P.O. Box 2240, Hail 55476, Saudi Arabia
| | - Samra Siddiqui
- Department
of Health Services Management, College of Public Health and Health
Informatics, University of Hail, Hail 55476, Saudi Arabia
| | - Mohammad Abohassan
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Snober S. Mir
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow 226026, India
| |
Collapse
|
4
|
Luo X. Nanobiotechnology-based strategies in alleviation of chemotherapy-mediated cardiotoxicity. ENVIRONMENTAL RESEARCH 2023; 238:116989. [PMID: 37633635 DOI: 10.1016/j.envres.2023.116989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
The cardiovascular diseases have been among the most common malignancies and the first leading cause of death, even higher than cancer. The cardiovascular diseases can be developed as a result of cardiac dysfunction and damages to heart tissue. Exposure to toxic agents and chemicals that induce cardiac dysfunction has been of interest in recent years. The chemotherapy drugs are commonly used for cancer therapy and in these patients, cardiovascular diseases have been widely observed that is due to negative impact of chemotherapy drugs on the heart. These drugs increase oxidative damage and inflammation, and mediate apoptosis and cardiac dysfunction. Hence, nanotechnological approaches have been emerged as new strategies in attenuation of chemotherapy-mediated cardiotoxicity. The first advantage of nanoparticles can be explored in targeted and selective delivery of drugs to reduce their accumulation in heart tissue. Nanostructures can deliver bioactive and therapeutic compounds in reducing cardiotoxicity and alleviation toxic impacts of chemotherapy drugs. The functionalization of nanostructures increases their selectivity against tumor cells and reduces accumulation of drugs in heart tissue. The bioplatforms such as chitosan and alginate nanostructures can also deliver chemotherapy drugs and reduce their cardiotoxicity. The function of nanostructures is versatile in reduction of cardiotoxicity by chemotherapy drugs and new kind of platforms is hydrogels that can mediate sustained release of drug to reduce its toxic impacts on heart tissue. The various kinds of nanoplatforms have been developed for alleviation of cardiotoxicity and their future clinical application depends on their biocompatibility. High concentration level of chitosan nanoparticles can stimulate cardiotoxicity. Therefore, if nanotechnology is going to be deployed for drug delivery and reducing cardiotoxicity, the first pre-requirement is to lack toxicity on normal cells and have high biocompatibility.
Collapse
Affiliation(s)
- Xuanming Luo
- Department of General Surgery, Zhongshan Hospital, Fudan University, China; Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China.
| |
Collapse
|
5
|
Mirzaei S, Khademi Z, Zolfaghari R, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM. Dual-targeted delivery system using hollow silica nanoparticles with H +-triggered bubble generating characteristic coated with hyaluronic acid and AS1411 for cancer therapy. Drug Dev Ind Pharm 2023; 49:648-657. [PMID: 37772892 DOI: 10.1080/03639045.2023.2265484] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 09/26/2023] [Indexed: 09/30/2023]
Abstract
OBJECTIVE Herein, a dual-targeting delivery system using mesoporous silica nanoparticles with hollow structures (HMSNs) was developed for the specific delivery of epirubicin (EPI) to cancer cells and introducing a H+-triggered bubble generating nanosystem (BGNS). HMSNs containing EPI are covered by hyaluronic acid (HA) shell and AS1411 aptamer to create the BGNS-EPI-HA-Apt complex, which is highly selective against CD44 marker and nucleolin overexpressed on the surface of tumor cells. METHODS MTT assay compared the cytotoxicity of different treatments in CHO (Chinese hamster ovary) cells as well as 4T1 (murine mammary carcinoma) and MCF-7 (human breast adenocarcinoma) cells. The internalization of Epi was assessed by flow cytometry along with fluorescence imaging. In vivo studies were conducted on BALB/c mice bearing a tumor from 4T1 cell line where monitoring included measuring tumor volume, mouse weight changes over time alongside mortality rate; accumulation levels for Epi within organs were also measured during this process. RESULTS The collected data illustrated that BGNS-EPI-HA-Apt complex controlled the release of EPI in a sustained method. Afterward, receptor-mediated internalization via nucleolin and CD44 was verified in 4T1 and MCF-7 cells using fluorescence microscopy assay and flow cytometry analysis. The results of tumor inhibitory effect study exhibited that BGNS-EPI-HA-Apt complex decreased off-target effect and improved on-target effects because of its targeting ability. CONCLUSION The data acquired substantiates that HA-surface modified HMSNs functionalized with aptamers possess significant potential as a focused platform for efficient transportation of anticancer agents to neoplastic tissues.
Collapse
Affiliation(s)
- Salimeh Mirzaei
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Zahra Khademi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Zolfaghari
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Shakir N, Sharif A, Ali S, Akhtar B, Akhtar MF, Muhammad F, Saleem A, Akhtar K, Tariq I, Khan MI. Pirarubicin loaded biodegradable nanoparticles downregulate IL-6, COX-II and TNF-α along with oxidative stress markers in comparison to conventional pirarubicin in healthy albino rats. J Drug Deliv Sci Technol 2023; 84:104498. [DOI: 10.1016/j.jddst.2023.104498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
|
7
|
Kesharwani P, Chadar R, Shukla R, Jain GK, Aggarwal G, Abourehab MAS, Sahebkar A. Recent advances in multifunctional dendrimer-based nanoprobes for breast cancer theranostics. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2433-2471. [PMID: 35848467 DOI: 10.1080/09205063.2022.2103627] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Breast cancer (BC) undoubtedly is one of the most common type of cancers amongst women, which causes about 5 million deaths annually. The treatments and diagnostic therapy choices currently available for Breast Cancer is very much limited . Advancements in novel nanocarrier could be a promising strategy for diagnosis and treatments of this deadly disease. Dendrimer nanoformulation could be functionalized and explored for efficient targeting of overexpressed receptors on Breast Cancer cells to achieve targeted drug delivery, for diagnostics and to overcome the resistance of the cells towards particular chemotherapeutic. Additionally, the dendrimer have shown promising potential in the improvement of therapeutic value for Breast Cancer therapy by achieving synergistic co-delivery of chemotherapeutics and genetic materials for multidirectional treatment. In this review, we have highlighted the application of dendrimer as novel multifunctional nanoplatforms for the treatment and diagnosis of Breast Cancer.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.,University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Rahul Chadar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P, India
| | - Gaurav K Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Neuroprotective Effect of Artichoke-Based Nanoformulation in Sporadic Alzheimer’s Disease Mouse Model: Focus on Antioxidant, Anti-Inflammatory, and Amyloidogenic Pathways. Pharmaceuticals (Basel) 2022; 15:ph15101202. [PMID: 36297313 PMCID: PMC9610800 DOI: 10.3390/ph15101202] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
The vast socio-economic impact of Alzheimer’s disease (AD) has prompted the search for new neuroprotective agents with good tolerability and safety profile. With its outstanding role as antioxidant and anti-inflammatory, alongside its anti-acetylcholinesterase activity, the artichoke can be implemented in a multi-targeted approach in AD therapy. Moreover, artichoke agricultural wastes can represent according to the current United Nations Sustainable Development goals an opportunity to produce medicinally valuable phenolic-rich extracts. In this context, the UPLC-ESI-MS/MS phytochemical characterization of artichoke bracts extract revealed the presence of mono- and di-caffeoylquinic acids and apigenin, luteolin, and kaempferol O-glycosides with remarkable total phenolics and flavonoids contents. A broad antioxidant spectrum was established in vitro. Artichoke-loaded, chitosan-coated, solid lipid nanoparticles (SLNs) were prepared and characterized for their size, zeta potential, morphology, entrapment efficiency, release, and ex vivo permeation and showed suitable colloidal characteristics, a controlled release profile, and promising ex vivo permeation, indicating possibly better physicochemical and biopharmaceutical parameters than free artichoke extract. The anti-Alzheimer potential of the extract and prepared SLNs was assessed in vivo in streptozotocin-induced sporadic Alzheimer mice. A great improvement in cognitive functions and spatial memory recovery, in addition to a marked reduction of the inflammatory biomarker TNF-α, β-amyloid, and tau protein levels, were observed. Significant neuroprotective efficacy in dentate Gyrus sub-regions was achieved in mice treated with free artichoke extract and to a significantly higher extent with artichoke-loaded SLNs. The results clarify the strong potential of artichoke bracts extract as a botanical anti-AD drug and will contribute to altering the future medicinal outlook of artichoke bracts previously regarded as agro-industrial waste.
Collapse
|
9
|
Ranasinghe R, Mathai M, Zulli A. Revisiting the therapeutic potential of tocotrienol. Biofactors 2022; 48:813-856. [PMID: 35719120 PMCID: PMC9544065 DOI: 10.1002/biof.1873] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
The therapeutic potential of the tocotrienol group stems from its nutraceutical properties as a dietary supplement. It is largely considered to be safe when consumed at low doses for attenuating pathophysiology as shown by animal models, in vitro assays, and ongoing human trials. Medical researchers and the allied sciences have experimented with tocotrienols for many decades, but its therapeutic potential was limited to adjuvant or concurrent treatment regimens. Recent studies have focused on targeted drug delivery by enhancing the bioavailability through carriers, self-sustained emulsions, nanoparticles, and ethosomes. Epigenetic modulation and computer remodeling are other means that will help increase chemosensitivity. This review will focus on the systemic intracellular anti-cancer, antioxidant, and anti-inflammatory mechanisms that are stimulated and/or regulated by tocotrienols while highlighting its potent therapeutic properties in a diverse group of clinical diseases.
Collapse
Affiliation(s)
- Ranmali Ranasinghe
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| | - Michael Mathai
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| | - Anthony Zulli
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| |
Collapse
|
10
|
Chakraborty E, Sarkar D. Emerging Therapies for Hepatocellular Carcinoma (HCC). Cancers (Basel) 2022; 14:2798. [PMID: 35681776 PMCID: PMC9179883 DOI: 10.3390/cancers14112798] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 01/30/2023] Open
Abstract
Hepatocellular carcinoma (HCC) arises from hepatocytes and accounts for 90% of primary liver cancer. According to Global Cancer Incidence, Mortality and Prevalence (GLOBOCAN) 2020, globally HCC is the sixth most common cancer and the third most common cause of cancer-related deaths. Reasons for HCC prognosis remaining dismal are that HCC is asymptomatic in its early stages, leading to late diagnosis, and it is markedly resistant to conventional chemo- and radiotherapy. Liver transplantation is the treatment of choice in early stages, while surgical resection, radiofrequency ablation (RFA) and trans arterial chemoembolization (TACE) are Food and Drug Administration (FDA)-approved treatments for advanced HCC. Additional first line therapy for advanced HCC includes broad-spectrum tyrosine kinase inhibitors (TKIs), such as sorafenib and lenvatinib, as well as a combination of immunotherapy and anti-angiogenesis therapy, namely atezolizumab and bevacizumab. However, these strategies provide nominal extension in the survival curve, cause broad spectrum toxic side effects, and patients eventually develop therapy resistance. Some common mutations in HCC, such as in telomerase reverse transcriptase (TERT), catenin beta 1 (CTNNB1) and tumor protein p53 (TP53) genes, are still considered to be undruggable. In this context, identification of appropriate gene targets and specific gene delivery approaches create the potential of gene- and immune-based therapies for the safe and effective treatment of HCC. This review elaborates on the current status of HCC treatment by focusing on potential gene targets and advanced techniques, such as oncolytic viral vectors, nanoparticles, chimeric antigen receptor (CAR)-T cells, immunotherapy, and clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9), and describes future prospects in HCC treatment.
Collapse
Affiliation(s)
- Eesha Chakraborty
- C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Massey Cancer Center, VCU Institute of Molecular Medicine (VIMM), Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
11
|
Zainal Z, Khaza'ai H, Kutty Radhakrishnan A, Chang SK. Therapeutic potential of palm oil vitamin E-derived tocotrienols in inflammation and chronic diseases: Evidence from preclinical and clinical studies. Food Res Int 2022; 156:111175. [DOI: 10.1016/j.foodres.2022.111175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/17/2022] [Accepted: 03/17/2022] [Indexed: 12/17/2022]
|
12
|
Yang Z, Deng W, Zhang X, An Y, Liu Y, Yao H, Zhang Z. Opportunities and Challenges of Nanoparticles in Digestive Tumours as Anti-Angiogenic Therapies. Front Oncol 2022; 11:789330. [PMID: 35083147 PMCID: PMC8784389 DOI: 10.3389/fonc.2021.789330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/10/2021] [Indexed: 01/04/2023] Open
Abstract
Digestive tumours, a common kind of malignancy worldwide, have recently led to the most tumour-related deaths. Angiogenesis, the process of forming novel blood vessels from pre-existing vessels, is involved in various physiological and pathological processes in the body. Many studies suggest that abnormal angiogenesis plays an important role in the growth, progression, and metastasis of digestive tumours. Therefore, anti-angiogenic therapy is considered a promising target for improving therapeutic efficacy. Traditional strategies such as bevacizumab and regorafenib can target and block the activity of proangiogenic factors to treat digestive tumours. However, due to resistance and some limitations, such as poor pharmacokinetics, their efficacy is not always satisfactory. In recent years, nanotechnology-based anti-angiogenic therapies have emerged as a new way to treat digestive tumours. Compared with commonly used drugs, nanoparticles show great potential in tumour targeted delivery, controlled drug release, prolonged cycle time, and increased drug bioavailability. Therefore, anti-angiogenic nanoparticles may be an effective complementary therapy to treat digestive tumours. In this review, we outline the different mechanisms of angiogenesis, the effects of nanoparticles on angiogenesis, and their biomedical applications in various kinds of digestive tumours. In addition, the opportunities and challenges are briefly discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongwei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
13
|
Liu F, Zhu XT, Li Y, Wang CJ, Fu JL, Hui J, Xiao Y, Liu L, Yan R, Li XF, Liu Y. Magnesium demethylcantharidate inhibits hepatocellular carcinoma cell invasion and metastasis via activation transcription factor FOXO1. Eur J Pharmacol 2021; 911:174558. [PMID: 34634308 DOI: 10.1016/j.ejphar.2021.174558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 11/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world, develops rapidly and has a high mortality rate. Relapsed metastasis is the most important factor affecting prognosis and is also the main cause of death for patients with HCC. Cantharidin is a kind of folk medicine for malignant tumors in China. Because of its cytotoxicity, the application of cantharidin is very limited. Magnesium demethylcantharidate (MDC) is a derivative of cantharidin independently developed by our laboratory. Our results show that MDC has anticancer activity and exhibited lower toxicity than cantharidin. However, whether MDC affects the invasion and metastasis of HCC cells and the underlying molecular mechanisms remain obscure. Transwell and Matrigel assays showed that MDC could effectively inhibit the invasion and metastasis of the HCC cell lines SMMC-7721 and SK-Hep1 in a dose-dependent manner. Moreover, MDC significantly inhibited the expression of invasion and metastasis related proteins MMP-2 and MMP-9. In addition, our study found that MDC inhibited the invasion and metastasis of HCC cell lines SMMC-7721 and SK-Hep1 by activating transcription factor FOXO1. Interestingly, the combination of MDC and sorafenib significantly inhibited the invasion and metastasis of HCC cell lines SMMC-7721 and SK-Hep1 compared with the single drug treatment via the activated transcription factor FOXO1. Our work revealed that MDC obviously inhibited the invasion and metastasis of HCC cells, and suggested that MDC could be a potential candidate molecule against the invasion and metastasis of HCC.
Collapse
Affiliation(s)
- Fang Liu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xin-Ting Zhu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China; Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China
| | - Yi Li
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Chen-Jing Wang
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Jia-Li Fu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Jing Hui
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China; Life Sciences Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Yi Xiao
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China; Life Sciences Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Liu Liu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Rong Yan
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xiao-Fei Li
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Yun Liu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China; Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China; Life Sciences Institute, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
14
|
He J, Sun H, Li F, Yang H, Lou M, Wang S, Wu C. Efficacy and safety of raltitrexed-based transcatheter arterial chemoembolization for intermediate and advanced hepatocellular carcinoma: A multicenter real-world study. Hepatol Res 2021; 51:1153-1163. [PMID: 34492152 DOI: 10.1111/hepr.13708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022]
Abstract
AIM This study aimed to evaluate efficacy and safety of raltitrexed-based transcatheter arterial chemoembolization (TACE) for intermediate and advanced hepatocellular carcinoma (HCC) using real-world evidence. METHODS All eligible HCC cases were collected from multiple centers in Chongqing, China, from January 2013 to December 2018 and divided into the raltitrexed group (raltitrexed + lobaplatin + pirarubicin) and control group (lobaplatin + pirarubicin). Propensity score matching (PSM) with a 1:1 ratio was used to eliminate the imbalance of potential confounding factors between groups. The primary end-point was overall survival (OS) and the secondary end-points were progression-free survival (PFS) and disease control rate. RESULTS The median follow-up period for patients in the raltitrexed and control groups was 8.7 and 5.9 months, respectively. After PSM, median OS was 10.0 months in the raltitrexed group and 7.0 months in the control group (p = 0.002). The 6-month, 1-year, and 2-year OS rates of the raltitrexed group were significantly higher than those of the control group (78.2% vs. 60.9%, p = 0.010; 43.5% vs. 22.8%, p = 0.030; and 17.4% vs. 2.2% p = 0.001, respectively). Multivariate analysis of these propensity score-matched HCC patients revealed treatment, age, tumor size, lipiodol accumulation, and the number of TACE cycles as independent predictors of OS (all p < 0.05). The disease control rate of the raltitrexed and control groups was 87.4% and 65.8%, respectively (p < 0.001). CONCLUSIONS Raltitrexed-based TACE can prolong the OS of patients with intermediate and advanced HCC in a real-world clinical setting, and is safe and tolerable.
Collapse
Affiliation(s)
- Jiahui He
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hang Sun
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fei Li
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Yang
- Department of Respiration, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Minggeng Lou
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shunde Wang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chuanxin Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Therapeutic strategies for miRNA delivery to reduce hepatocellular carcinoma. Semin Cell Dev Biol 2021; 124:134-144. [PMID: 33926792 DOI: 10.1016/j.semcdb.2021.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/21/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022]
Abstract
Malignancies of hepatocellular carcinoma (HCC) are rapidly spreading and commonly fatal. Like most cancers, the gene expression patterns in HCC vary significantly from patient to patient. Moreover, the expression networks during HCC progression are largely controlled by microRNAs (miRNAs) regulating multiple oncogenes and tumor supressors. Therefore, miRNA-based therapeutic strategies altering these networks may significantly influence the cellular behavior enough for them to cure HCC. However, the most substantial challenges in developing such therapies are the stability of the oligos themselves and that of their delivery systems. Here we provide a comprehensive update describing various miRNA delivery systems, including virus-based delivery and non-viral delivery. The latter may be achieved using inorganic nanoparticles, polymer based nano-carriers, lipid-based vesicles, exosomes, and liposomes. Leaky vasculature in HCC-afflicted livers helps untargeted nanocarriers to accumulate in the tumor tissue but may result in side effects during higher dose of treatment. On the other hand, the strategies for actively targeting miRNA therepeutics to cancerous cells through nano-conjugates or vesicles by decorating their surface with antibodies against or ligands for HCC-specific antigens or receptors are more efficient in preventing damage to healthy tissue and cancer recurrence.
Collapse
|
16
|
Folic acid decorated chitosan-coated solid lipid nanoparticles for the oral treatment of rheumatoid arthritis. Ther Deliv 2021; 12:297-310. [PMID: 33726498 DOI: 10.4155/tde-2020-0123] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Systemic treatment of rheumatoid arthritis has been accompanied with several side effects. This study attempts to reduce leflunomide systemic side effects besides increasing its joint healing outcomes via formulation of layer-by-layer coated, leflunomide-loaded solid lipid nanoparticles (SLNs). Methods: SLNs were coated with chitosan (CS) followed by folic acid (FA). FA-CS-SLNs were about 284.9 nm and carried negative surface charge. Results & conclusion: FA-CS-SLNs showed sustained release profile for 168 h. Results of oral administration of FA-CS-SLNs in rats with adjuvant-induced arthritis revealed improved joint healing and reduced hepatotoxicity compared with leflunomide suspension. This may be attributed to the ability of FA-CS-SLNs to actively target FA receptors that are overexpressed in inflamed rheumatic joints in addition to innate joint healing properties of CS.
Collapse
|
17
|
Chiu HI, Samad NA, Fang L, Lim V. Cytotoxicity of targeted PLGA nanoparticles: a systematic review. RSC Adv 2021; 11:9433-9449. [PMID: 35423427 PMCID: PMC8695459 DOI: 10.1039/d1ra00074h] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Recent advances in nanotechnology have contributed tremendously to the development and revolutionizing of drug delivery systems in the field of nanomedicine. In particular, targeting nanoparticles based on biodegradable poly(lactic-co-glycolic acid) (PLGA) polymers have gained much interest. However, PLGA nanoparticles remain of concern for their effectiveness against cancer cells and their toxicity to normal cells. The aim of this systematic review is to identify a promising targeting PLGA nanoformulation based on the comparison study of their cytotoxicity potency in different cell lines. A literature search was conducted through the databases of Google Scholar, PubMed, ScienceDirect, Scopus and SpringerLink. The sources studied were published between 2009 and 2019, and a variety of keywords were utilized. In total, 81 manuscripts that met the inclusion and exclusion criteria were selected for analysis based on their cytotoxicity, size, zeta potential, year of publication, type of ligand, active compounds and cell line used. The half maximal inhibitory concentration (IC50) for cytotoxicity was the main measurement in this data extraction, and the SI units were standardized to μg mL-1 for a better view of comparison. This systematic review also identified that cytotoxicity potency was inversely proportional to nanoparticle size. The PLGA nanoparticles predominantly exhibited a size of less than 300 nm and absolute zeta potential ∼20 mV. In conclusion, more comprehensive and critical appraisals of pharmacokinetic, pharmacokinetic, toxicokinetic, in vivo and in vitro tests are required for the investigation of the full value of targeting PLGA nanoparticles for cancer treatment.
Collapse
Affiliation(s)
- Hock Ing Chiu
- Integrative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia Bertam 13200 Kepala Batas Penang Malaysia +604-5622427
| | - Nozlena Abdul Samad
- Integrative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia Bertam 13200 Kepala Batas Penang Malaysia +604-5622427
| | - Lizhen Fang
- School of Pharmacy, Xinxiang Medical University Xinxiang Henan 453003 People's Republic of China
| | - Vuanghao Lim
- Integrative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia Bertam 13200 Kepala Batas Penang Malaysia +604-5622427
| |
Collapse
|
18
|
Kumar V, Rahman M, Gahtori P, Al-Abbasi F, Anwar F, Kim HS. Current status and future directions of hepatocellular carcinoma-targeted nanoparticles and nanomedicine. Expert Opin Drug Deliv 2020; 18:673-694. [PMID: 33295218 DOI: 10.1080/17425247.2021.1860939] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a major health problem worldwide. Conventional therapies covering either chemotherapy or combination therapy still have sub-optimal responses with significant adverse effects and toxicity. Moreover, tumor cells usually acquire resistance quickly for traditional approaches, limiting their use in HCC. Interest in nanomedicine due to minimal systemic toxicity and a high degree of target-specific drug-delivery have pulled the attention of health scientists in this area of therapeutics. AREA COVERED The review covers the incidence and epidemiology of HCC, proposed molecular drug targets, mechanistic approach and emergence of nanomedicines including nanoparticles, lipidic nanoparticles, vesicular-based nanocarrier, virus-like particles with momentous therapeutic aspects including biocompatibility, and toxicity of nanocarriers along with conclusions and future perspective, with an efficient approach to safely cross physiological barriers to reach the target site for treating liver cancer. EXPERT OPINION Remarkable outcomes have recently been observed for the therapeutic efficacy of nanocarriers with respect to a specific drug target against the treatment of HCC by existing under trial drugs.
Collapse
Affiliation(s)
- Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Mahfoozur Rahman
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Prashant Gahtori
- School of Pharmacy, Graphic Era Hill University Dehradun 248002, Uttarakhand, India
| | - Fahad Al-Abbasi
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Firoz Anwar
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Jangan-gu, Suwon 16419, 2066, Seobu-ro, Korea
| |
Collapse
|
19
|
Abo Mansour HE, El-Batsh MM, Badawy NS, Mehanna ET, Mesbah NM, Abo-Elmatty DM. Ginger Extract Loaded into Chitosan Nanoparticles Enhances Cytotoxicity and Reduces Cardiotoxicity of Doxorubicin in Hepatocellular Carcinoma in Mice. Nutr Cancer 2020; 73:2347-2362. [PMID: 32972241 DOI: 10.1080/01635581.2020.1823436] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/27/2020] [Accepted: 09/05/2020] [Indexed: 02/08/2023]
Abstract
This study aimed to investigate the impact of ginger extract (GE) loaded into chitosan nanoparticles (CNPs) in enhancing cytotoxicity and reducing cardiotoxicity of doxorubicin (DXN) in hepatocellular carcinoma (HCC) induced mice. DXN and GE were loaded into CNPs and cytotoxicity of loaded and unloaded drugs against HepG2 cells was evaluated. HCC was induced in male albino mice by injection of diethylnitrosamine (DINA). Mice were divided into eight groups (n = 15): (1) normal control, (2) DINA, (3) CNPs, (4) free DXN, (5) CNPs DXN, (6) free GE, (7) CNPs GE, and (8) CNPs DXN + CNPs GE. Both GE and DXN loaded into CNPs showed a greater decline in cell viability of HepG2 cells than the unloaded forms. GE CNPs displayed pronounced anticancer activity In Vivo through apoptosis, greater down-regulation of multidrug resistance 1, enhancement of anti-oxidant activity and depletion of vascular endothelial growth factor content in liver tissues. GE CNPs in combination with DXN CNPs showed nearly normal hepatic lobule architecture and the greatest increase in apoptotic cell count. Co-treatment group had decreased cardiac malondialdehyde, tumor necrosis factor-α and serum activity of creatine kinase and lactate dehydrogenase. Combination of GE CNPs and DXN CNPs might be a potentially effective therapeutic approach for HCC.
Collapse
Affiliation(s)
- Hend E Abo Mansour
- Department of Biochemistry, Faculty of Pharmacy, Menoufia University, Shebeen El-Kom, Egypt
| | - Maha M El-Batsh
- Department of Clinical Pharmacology, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Nadia S Badawy
- Department of Histology and Cell Biology, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Noha M Mesbah
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Dina M Abo-Elmatty
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
20
|
Xiao Y, Gong Q, Wang W, Liu F, Kong Q, Pan F, Zhang X, Yu C, Hu S, Fan F, Li S, Liu Y. The combination of Biochanin A and SB590885 potentiates the inhibition of tumour progression in hepatocellular carcinoma. Cancer Cell Int 2020; 20:371. [PMID: 32774165 PMCID: PMC7405455 DOI: 10.1186/s12935-020-01463-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most aggressive and frequently diagnosed malignancy of the liver. Despite aggressive therapy, life expectancy of many patients in these cases is extended by only a few months. Hepatocellular carcinoma (HCC) has a particularly poor prognosis and would greatly benefit from more effective therapies. Methods The CCK-8 assay and colony formation assays were used to test the cell proliferation and viability. The effects of combination Biochanin A and SB590885 on apoptosis and cell cycle arrest of HCC cells were analysed by flow cytometry. The expression of ERK MAPK and PI3K/AKT/mTOR signalling as well as apoptosis and cell cycle-related proteins in HCC cells were tested by western blotting. The HCC cell xenograft model was established to test the tumor proliferation. Serum and plasma were tested for liver and kidney safety markers (ALP, ALT, AST, total bilirubin, creatinine, urea nitrogen) by using SpectraMax i3X. Results The combination of natural product Biochanin A with the BRAF inhibitor SB590885 synergistically suppressed proliferation, and promoted cell cycle arrest and apoptosis in vitro. Furthermore, we demonstrated that the combination of Biochanin A and SB590885 led to increased impairment of proliferation and HCC tumour inhibition through disrupting of the ERK MAPK and the PI3K/AKT pathways in vitro. The volumes tumors and the weights of tumours were significantly reduced by the combination treatment compared to the control or single treatments in vivo. In addition, we found that there was no significant hepatorenal toxicity with the drug combination, as indicated by the hepatorenal toxicity test. Conclusion The results identify an effective combination therapy for the most aggressive form of HCC and provide the possibility of therapeutic improvement for patients with advanced HCC.
Collapse
Affiliation(s)
- Yi Xiao
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Department of Biochemistry and Molecular Biology, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Qiang Gong
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Wenhong Wang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Fang Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China
| | - Qinghong Kong
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Feng Pan
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Xiaoke Zhang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Changyan Yu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Shanshan Hu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 China
| | - Fang Fan
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Department of Biochemistry and Molecular Biology, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China
| | - Sanhua Li
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Yun Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Department of Biochemistry and Molecular Biology, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| |
Collapse
|
21
|
Understanding fundamentals of hepatocellular carcinoma to design next-generation chitosan nano-formulations: Beyond chemotherapy stride. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
22
|
Abo Mansour HE, El-Batsh MM, Badawy NS, Mehanna ET, Mesbah NM, Abo-Elmatty DM. Effect of co-treatment with doxorubicin and verapamil loaded into chitosan nanoparticles on diethylnitrosamine-induced hepatocellular carcinoma in mice. Hum Exp Toxicol 2020; 39:1528-1544. [PMID: 32519553 DOI: 10.1177/0960327120930266] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This study aimed to investigate the potential role of co-treatment with doxorubicin (DOX) and verapamil (VRP) nanoparticles in experimentally induced hepatocellular carcinoma in mice and to investigate the possible mechanisms behind the potential favorable effect of the co-treatment. DOX and VRP were loaded into chitosan nanoparticles (CHNPs), and cytotoxicity of loaded and unloaded drugs against HepG2 cells was evaluated. Male albino mice were divided into eight groups (n = 15): (1) normal control, (2) diethylnitrosamine, (3) CHNPs, (4) free DOX, (5) CHNPs DOX, (6) free VRP, (7) CHNPs VRP, and (8) CHNPs DOX + CHNPs VRP. Either VRP or DOX loaded into CHNPs showed stronger growth inhibition of HepG2 cells than their free forms. DOX or VRP nanoparticles displayed pronounced anticancer activity in vivo through the decline of vascular endothelial growth factor and B cell lymphoma-2 contents in liver tissues, upregulation of antioxidant enzymes, and downregulation of multidrug resistance 1. Moreover, reduced cardiotoxicity was evident from decreased level of tumor necrosis factor-α and malondialdehyde in heart tissues coupled with decreased serum activity of creatine kinase-myocardial band and lactate dehydrogenase. Co-treatment with CHNPs DOX and CHNPs VRP showed superior results versus other treatments. Liver sections from the co-treatment group revealed the absence of necrosis, enhanced apoptosis, and nearly normal hepatic lobule architecture. Co-treatment with CHNPs DOX and CHNPs VRP revealed enhanced anticancer activity and decreased cardiotoxicity versus the corresponding free forms.
Collapse
Affiliation(s)
- H E Abo Mansour
- Department of Biochemistry, Faculty of Pharmacy, Menoufia University, Shibin el Kom, Egypt
| | - M M El-Batsh
- Department of Clinical Pharmacology, Faculty of Medicine, Menoufia University, Shibin el Kom, Egypt
| | - N S Badawy
- Department of Histology and Cell Biology, Faculty of Medicine, Menoufia University, Shibin el Kom, Egypt
| | - E T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - N M Mesbah
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - D M Abo-Elmatty
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
23
|
Chitosan Nanoparticles for Therapy and Theranostics of Hepatocellular Carcinoma (HCC) and Liver-Targeting. NANOMATERIALS 2020; 10:nano10050870. [PMID: 32365938 PMCID: PMC7279387 DOI: 10.3390/nano10050870] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/24/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022]
Abstract
Chitosan nanoparticles are well-known delivery systems widely used as polymeric carriers in the field of nanomedicine. Chitosan is a carbohydrate of natural origin: it is a biodegradable, biocompatible, mucoadhesive, polycationic polymer and it is endowed with penetration enhancer properties. Furthermore, it can be easily derivatized. Hepatocellular carcinoma (HCC) represents a remarkable health problem because current therapies, that include surgery, liver transplantation, trans-arterial embolization, chemoembolization and chemotherapy, present significant limitations due to the high risk of recurrence, to a lack of drug selectivity and to other serious side effects. Therefore, there is the need for new therapeutic strategies and for improving the liver-targeting to HCC. Nanomedicine consists in the use of nanoscale carriers as delivery systems to target and deliver drugs and/or diagnostic agents to specific organs or tissues. Chitosan and its derivatives can be successfully used in the preparation of nanoparticles that, for their peculiar surface-properties, can specifically interact with liver tumor, by passive and active targeting. This review concerns the use of chitosan nanoparticles for the therapy and theranostics of HCC and liver-targeting.
Collapse
|
24
|
El-Sisi AE, Sokkar SS, Ibrahim HA, Hamed MF, Abu-Risha SE. Targeting MDR-1 gene expression, BAX/BCL2, caspase-3, and Ki-67 by nanoencapsulated imatinib and hesperidin to enhance anticancer activity and ameliorate cardiotoxicity. Fundam Clin Pharmacol 2020; 34:458-475. [PMID: 32080901 DOI: 10.1111/fcp.12549] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 02/08/2020] [Accepted: 02/19/2020] [Indexed: 12/21/2022]
Abstract
There is a great demand to introduce new approaches into cancer treatment field due to incidence of increased breast cancer all over the world. The current study was designed to evaluate the role of imatinib mesylate (IM) and/or hesperidin (HES) nanoparticles alone or in combination in enhancing the anticancer activity and to investigate the ability of nanoencapsulation to reduce cardiotoxicity of IM in solid Ehrlich carcinoma (SEC)-bearing mice. IM and HES were loaded into PLGA (poly(lactic-co-glycolic acid) polymer. SEC was induced in female albino mice as a model for experimentally induced breast cancer. Mice were randomly divided into eight groups (n = 10). On day 28 from tumor inoculation, mice were sacrificed and blood samples were collected in heparinized tubes for hematological studies, biochemical determination of lactate dehydrogenase (LDH), and glutamic oxaloacetic transaminase (SGOT) levels. In addition, tumor and cardiac tissues were utilized for histopathological examination as well as determination of MDR-1 gene expression. Immunohistochemical staining of BAX and BCL-2 was done. Nano IM- and/or Nano HES-treated groups showed a significant reduction in tumor volume, weight, hematological, cardiac markers, and tumor MDR-1 gene downregulation compared to free conventional treated groups. In conclusion, the use of HES as an adjuvant therapy with IM could improve its cytotoxic effects and limit its cardiac toxicity. Furthermore, nanoencapsulation of IM and/or HES with PLGA polymer showed a remarkable anticancer activity.
Collapse
Affiliation(s)
- Alaa E El-Sisi
- Pharmacology and Toxicology department, College of Pharmacy, University of Tanta, Tanta, Egypt
| | - Samia S Sokkar
- Pharmacology and Toxicology department, College of Pharmacy, University of Tanta, Tanta, Egypt
| | - Hanaa A Ibrahim
- Pharmacology and Toxicology department, College of Pharmacy, University of Tanta, Tanta, Egypt
| | - Mohamed F Hamed
- Department of Pathology, College of Veterinary Medicine, University of El-Mansoura, Mansoura, Egypt
| | - Sally E Abu-Risha
- Pharmacology and Toxicology department, College of Pharmacy, University of Tanta, Tanta, Egypt
| |
Collapse
|
25
|
Cacan E, Ozmen ZC. Regulation of Fas in response to bortezomib and epirubicin in colorectal cancer cells. J Chemother 2020; 32:193-201. [PMID: 32162602 DOI: 10.1080/1120009x.2020.1740389] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Bortezomib is a reversible proteasome inhibitor affects the ubiquitin-proteasome mechanism to kill cancer cells, and inhibition of the proteasome modulates the expression of multiple target genes at the transcriptional level. Epirubicin is known as an anthracycline agent that interferes with DNA and RNA synthesis, and it can be used with other chemotherapeutic drugs in the treatment of post-surgical breast cancer. Epirubicin may have an anti-tumor effect against broad-spectrum tumor cells. However, it is a non-specific chemotherapeutic agent that can cause high toxicity if not used in appropriate doses. Here, we hypothesize that a combination treatment of bortezomib and epirubicin will induce immunogenic cell death in colorectal cancer cells by increasing expression of death receptors such as Fas, which will make these cancer cells more susceptible to Fas/FasL mediated tumor cell killing. Our data demonstrate that a combination of bortezomib and epirubicin significantly increases the sensitivity of colorectal carcinoma cells, but not healthy non-malignant epithelial cells, to apoptosis. The combination treatment significantly upregulates the transcriptional activation of Fas in colorectal cancer cells but not in normal cells. Our results suggest that combining bortezomib and epirubicin may simultaneously enhance tumor immunogenicity and the induction of antitumor immunity.
Collapse
Affiliation(s)
- Ercan Cacan
- Department of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Turkey
| | - Zeliha C Ozmen
- Department of Biochemistry, Tokat Gaziosmanpasa University, Tokat, Turkey
| |
Collapse
|
26
|
|
27
|
Erdogan A, Ozkan A. Cetuximab and epirubicin HCl-combined application as a possibility to treat both parental and epirubicin HCl-resistant liver cancer cells. Biol Futur 2019; 70:175-184. [PMID: 34554450 DOI: 10.1556/019.70.2019.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 07/17/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Targeted chemotherapeutics such as cetuximab can cause many side effects such as skin toxicity when used in high concentrations. In addition, cancer cells can develop resistance to some of the anticancer agents during treatment. The lack of the desired success in chemotherapy and the development of resistance to chemotherapeutics, such as epirubicin HCl, suggest that there is a need for combined therapies. The combination of targeted chemotherapeutics and conventional chemotherapy drugs may lead to the emergence of new strategies in the treatment of cancer. In this study, cytotoxic, antiproliferative, cell cycle inhibitive, oxidative stress generation, and apoptotic effects and effect mechanisms of cetuximab alone and together with epirubicin HCl on parental liver cancer cells (P-Hep G2) and epirubicin HCl-resistant liver cancer cells (R-Hep G2) were investigated. MATERIALS Cytotoxic effects of cetuximab alone and with epirubicin-HCl on cells were determined by Cell Titer-Blue® Cell Viability and Lactate Dehydrogenase Activity tests. Cell cycle distributions and apoptosis were detected by reverse transcription polymerase chain reaction (RT-PCR). RESULTS Cetuximab with epirubicin HCl treatment increased the cytotoxic effect on both cells. Caspase-3/7 activity increased 3 and 1.5 times in comparison with control group in P-Hep G2 and R-Hep G2 cells, respectively, after treating with cetuximab alone, whereas the increase was found to be approximately 4.7 and 2.5 times when cetuximab was treated with epirubicin HCl in P-Hep G2 and R-Hep G2 cells, respectively. Both cetuximab alone and together with epirubicin HCl treatments caused increases in Bax/Bcl-2 ratio in both cells. DISCUSSION Treatment of cetuximab with epirubicin HCl to P-Hep G2 and R-Hep G2 cells was found to be more effective in cytotoxic effect and inducing apoptosis comparison to cetuximab alone treatment. In addition, combination treatment showed different effects on pro-apoptotic/anti-apoptotic genes expression according to cells drug resistance properties.
Collapse
Affiliation(s)
- Ayse Erdogan
- Genetic and Bioengineering Department, Faculty of Engineering, Alanya Alaaddin Keykubat University, Antalya, Turkey.
| | - Aysun Ozkan
- Department of Biology, Faculty of Science, Akdeniz University, Antalya, Turkey
| |
Collapse
|
28
|
Zewail M, Nafee N, Helmy MW, Boraie N. Coated nanostructured lipid carriers targeting the joints – An effective and safe approach for the oral management of rheumatoid arthritis. Int J Pharm 2019; 567:118447. [DOI: 10.1016/j.ijpharm.2019.118447] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/02/2019] [Accepted: 06/17/2019] [Indexed: 12/16/2022]
|
29
|
Samie HAA, Saeed M, Faisal SM, Kausar MA, Kamal MA. Recent Findings on Nanotechnology-based Therapeutic Strategies Against Hepatocellular Carcinoma. Curr Drug Metab 2019; 20:283-291. [DOI: 10.2174/1389200220666190308134351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 12/14/2018] [Accepted: 02/14/2019] [Indexed: 02/07/2023]
Abstract
Background:
Nanotechnology-based therapies are emerging as a promising new anticancer approach.
Early clinical studies suggest that nanoparticle-based therapeutics can show enhanced efficacy while reducing side
effects minimal, owing to targeted delivery and active intracellular uptake.
Methods:
To overcome the problems of gene and drug delivery, nanotechnology based delivery system gained interest
in the last two decades. Encouraging results from Nano formulation based drug delivery systems revealed that
these emerging restoratives can efficiently lead to more effective, targeted, selective and efficacious delivery of chemotherapeutic
agents to the affected target cells.
Results:
Nanotechnology not only inhibits targeted gene products in patients with cancer, but also taught us valuable
lessons regarding appropriate dosages and route of administrations. Besides, nanotechnology based therapeutics
holds remarkable potential as an effective drug delivery system. We critically highlight the recent findings on
nanotechnology mediated therapeutics strategies to combat hepatocellular carcinoma and discuss how nanotechnology
platform can have enhanced anticancer effects compared with the parent therapeutic agents they contain.
Conclusion:
In this review, we discussed the key challenges, recent findings and future perspective in the development
of effective nanotechnology-based cancer therapeutics. The emphasis here is focused on nanotechnology-based
therapies that are likely to affect clinical investigations and their implications for advancing the treatment of patients
with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hany A. Abdel Samie
- Department of Zoology, Faculty of Science, Menoufia University, Al Minufya, Egypt
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Syed Mohd Faisal
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh-202002, India
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Hail, Saudi Arabia
| | - Mohammad A. Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
30
|
Zhu J, Xu J, Jiang LL, Huang JQ, Yan JY, Chen YW, Yang Q. Improved antitumor activity of cisplatin combined with Ganoderma lucidum polysaccharides in U14 cervical carcinoma-bearing mice. Kaohsiung J Med Sci 2019; 35:222-229. [PMID: 30958641 DOI: 10.1002/kjm2.12020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/20/2019] [Indexed: 02/01/2023] Open
Abstract
Research on anticervical cancer is urgently required to enhance clinical outcomes. As a main anticancer drug for cervical carcinoma, cisplatin (CIS) has been used for a lot of years in clinical therapy. However, serious adverse effects including nephrotoxicity and neurotoxicity limit its long-term treatment. Our main goal of this study is to investigate the improvement of Ganoderma lucidum polysaccharides (GPS) on CIS-induced antitumor effect of in U14 cervical carcinoma-bearing mice. The results showed that GPS + CIS could not only inhibit the growth of the tumor but also improve the spleen and thymus indexes. Moreover, little toxicological effects were observed on hepatic function and renal function in GPS + CIS treated mice bearing U14 tumor cells. Further analysis of the tumor inhibition mechanism indicated that the number of apoptotic tumor cells increased significantly, the expression of Bax increased and the expression of Bcl-2 decreased dramatically in cervical cancer sections after oral administration of GPS + CIS for 14 days. This GPS/CIS combined therapy represents intriguing therapeutic strategy for U14 cervical carcinoma providing not only superior efficacy but also a higher safety level.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, China
| | - Jia Xu
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, China
| | - Ling-Ling Jiang
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, China
| | - Jin-Qun Huang
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, China
| | - Jin-Yu Yan
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, China
| | - Yi-Wan Chen
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, China
| | - Qian Yang
- Department of Gynaecology and Obstetrics, First People's Hospital of Wenling, Wenling, China
| |
Collapse
|
31
|
Utilization of Vitamin E Analogs to Protect Normal Tissues While Enhancing Antitumor Effects. Semin Radiat Oncol 2019; 29:55-61. [PMID: 30573184 DOI: 10.1016/j.semradonc.2018.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite advances in radiation delivery techniques, side effects of radiation therapy due to radiation exposure of normal tissues are common and can limit the deliverable dose to tumors. Significant interests lie in pharmacologic modifiers that may protect against normal tissue toxicity from cancer treatment while simultaneously enhancing the tumor response to therapy. While no such treatments are available in the clinic, this is an area of active preclinical and clinical research. This review summarizes research studies that provide evidence to indicate that tocotrienols, natural forms of vitamin E, are potent radiation protectors and may also have antitumor effects. Hence, several current clinical trials test tocotrienols as concomitant treatment in cancer therapies.
Collapse
|
32
|
Influence of pravastatin chitosan nanoparticles on erythrocytes cholesterol and redox homeostasis: An in vitro study. ARAB J CHEM 2018. [DOI: 10.1016/j.arabjc.2015.10.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
33
|
Abdelmoneem MA, Mahmoud M, Zaky A, Helmy MW, Sallam M, Fang JY, Elkhodairy KA, Elzoghby AO. Decorating protein nanospheres with lactoferrin enhances oral COX-2 inhibitor/herbal therapy of hepatocellular carcinoma. Nanomedicine (Lond) 2018; 13:2377-2395. [DOI: 10.2217/nnm-2018-0134] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Lactoferrin (LF)-targeted gliadin nanoparticles (GL-NPs) were developed for targeted oral therapy of hepatocellular carcinoma. Materials & methods: Celecoxib and diosmin were incorporated in the hydrophobic matrix of GL-NPs whose surface was decorated with LF by electrostatic interaction for binding to asialoglycoprotein receptors overexpressed by liver cancer cells. Results: Targeted GL-NPs showed enhanced cytotoxic activity and increased cellular uptake in liver tumor cells compared with nontargeted NPs. Moreover, they demonstrated superior in vivo antitumor effects including reduction in the expression levels of tumor biomarkers and induction of caspase-mediated apoptosis. Ex vivo imaging of isolated organs exhibited extensive accumulation of NPs in livers more than other organs. Conclusion: LF-targeted GL-NPs could be considered as an efficient nanoplatform for targeted oral drug delivery for liver cancer therapy.
Collapse
Affiliation(s)
- Mona A Abdelmoneem
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mazen Mahmoud
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Amira Zaky
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Maged W Helmy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Damanhur University, Damanhur 22511, Egypt
| | - Marwa Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan
- Research Center for Industry of Human Ecology & Research Center for Chinese Herbal Medicine, Chang Gung University of Science & Technology, Kweishan, Taoyuan 333, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan 333, Taiwan
| | - Kadria A Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Division of Engineering in Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technologies, Cambridge, MA 02139, USA
| |
Collapse
|
34
|
Dual-targeted casein micelles as green nanomedicine for synergistic phytotherapy of hepatocellular carcinoma. J Control Release 2018; 287:78-93. [PMID: 30138716 DOI: 10.1016/j.jconrel.2018.08.026] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 07/22/2018] [Accepted: 08/16/2018] [Indexed: 12/29/2022]
Abstract
In recent years, green nanomedicines have made transformative difference in cancer therapy researches. Herein, we propose dual-functionalized spray-dried casein micelles (CAS-MCs) for combined delivery of two phytochemicals; berberine (BRB) and diosmin (DSN) as targeted therapy of hepatocellular carcinoma (HCC). The nanomicelles enabled parenteral delivery of the poorly soluble DSN via its encapsulation within their hydrophobic core. Moreover, sustained release of the water soluble BRB was attained by hydrophobic ion pairing with sodium deoxycholate followed by genipin crosslinking of CAS-MCs. Dual-active targeting of MCs, via conjugating both lactobionic acid (LA) and folic acid (FA), resulted in superior cytotoxicity and higher cellular uptake against HepG2 cells compared to single-targeted and non-targeted CAS-MCs. The dual-targeted DSN/BRB-loaded CAS-MCs demonstrated superior in vivo anti-tumor efficacy in HCC bearing mice as revealed by down regulation of cell necrosis markers (NF-κB and TNF-α), inflammatory marker COX2, inhibition of angiogenesis and induction of apoptosis. Histopathological analysis and immunohistochemical Ki67 staining confirmed the superiority of the dual-targeted micelles. Ex-vivo imaging showed preferential liver-specific accumulation of dual-targeted CAS-MCs. Overall, this approach combined the benefits of traditional herbal medicine with nanotechnology via LA/FA-CAS-MCs loaded with BRB and DSN as a promising nanoplatform for targeted HCC therapy.
Collapse
|
35
|
Liu XY, Zhou CB, Fang C. Nanomaterial-involved neural stem cell research: Disease treatment, cell labeling, and growth regulation. Biomed Pharmacother 2018; 107:583-597. [PMID: 30114642 DOI: 10.1016/j.biopha.2018.08.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) have been widely investigated for their potential in the treatment of various diseases and transplantation therapy. However, NSC growth regulation, labeling, and its application to disease diagnosis and treatment are outstanding challenges. Recently, nanomaterials have shown promise for various applications including genetic modification, imaging, and controlled drug release. Here we summarize the recent progress in the use of nanomaterials in combination with NSCs for disease treatment and diagnosis, cell labeling, and NSC growth regulation. The toxicity of nanomaterials to NSCs is also discussed.
Collapse
Affiliation(s)
- Xiang-Yu Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Cheng-Bin Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240 China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
36
|
Tocotrienols: The promising analogues of vitamin E for cancer therapeutics. Pharmacol Res 2018; 130:259-272. [DOI: 10.1016/j.phrs.2018.02.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/06/2018] [Accepted: 02/12/2018] [Indexed: 12/16/2022]
|
37
|
Lu J, Wang J, Ling D. Surface Engineering of Nanoparticles for Targeted Delivery to Hepatocellular Carcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:1702037. [PMID: 29251419 DOI: 10.1002/smll.201702037] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/24/2017] [Indexed: 05/20/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-associated deaths worldwide. There is a lack of efficient therapy for HCC; the only available first-line systemic drug, sorafenib, can merely improve the average survival by two months. Among the efforts to develop an efficient therapy for HCC, nanomedicine has drawn the most attention, owing to its unique features such as high drug-loading capacity, intrinsic anticancer activities, integrated diagnostic and therapeutic functionalities, and easy surface engineering with targeting ligands. Despite its tremendous advantages, no nanomedicine can be effective unless it successfully targets the tumor site, which is a challenging task. In this review, the features of HCC are described, and the physiological hurdles that prevent nanoparticles from targeting HCC are discussed. Then, the surface physicochemical factors of nanoparticles that can influence targeting efficiency are discussed. Finally, a thorough description of the physiological barriers that nanomedicine must conquer before uptake by HCC cells if possible is provided, as well as the surface engineering approaches to nanomedicine to achieve targeted delivery to HCC cells. The physiological hurdles and corresponding solutions summarized in this review provide a general guide for the rational design of HCC targeting nanomedicine systems.
Collapse
Affiliation(s)
- Jingxiong Lu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, and Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, 310058, China
| | - Jin Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, and Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, 310058, China
| | - Daishun Ling
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, and Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
38
|
Husain K, Malafa MP. Role of Tocotrienols in Chemosensitization of Cancer. ROLE OF NUTRACEUTICALS IN CHEMORESISTANCE TO CANCER 2018:77-97. [DOI: 10.1016/b978-0-12-812373-7.00004-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
39
|
Zheng P, Li J, Kros JM. Breakthroughs in modern cancer therapy and elusive cardiotoxicity: Critical research-practice gaps, challenges, and insights. Med Res Rev 2018; 38:325-376. [PMID: 28862319 PMCID: PMC5763363 DOI: 10.1002/med.21463] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 12/16/2022]
Abstract
To date, five cancer treatment modalities have been defined. The three traditional modalities of cancer treatment are surgery, radiotherapy, and conventional chemotherapy, and the two modern modalities include molecularly targeted therapy (the fourth modality) and immunotherapy (the fifth modality). The cardiotoxicity associated with conventional chemotherapy and radiotherapy is well known. Similar adverse cardiac events are resurging with the fourth modality. Aside from the conventional and newer targeted agents, even the most newly developed, immune-based therapeutic modalities of anticancer treatment (the fifth modality), e.g., immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapy, have unfortunately led to potentially lethal cardiotoxicity in patients. Cardiac complications represent unresolved and potentially life-threatening conditions in cancer survivors, while effective clinical management remains quite challenging. As a consequence, morbidity and mortality related to cardiac complications now threaten to offset some favorable benefits of modern cancer treatments in cancer-related survival, regardless of the oncologic prognosis. This review focuses on identifying critical research-practice gaps, addressing real-world challenges and pinpointing real-time insights in general terms under the context of clinical cardiotoxicity induced by the fourth and fifth modalities of cancer treatment. The information ranges from basic science to clinical management in the field of cardio-oncology and crosses the interface between oncology and onco-pharmacology. The complexity of the ongoing clinical problem is addressed at different levels. A better understanding of these research-practice gaps may advance research initiatives on the development of mechanism-based diagnoses and treatments for the effective clinical management of cardiotoxicity.
Collapse
Affiliation(s)
- Ping‐Pin Zheng
- Cardio‐Oncology Research GroupErasmus Medical CenterRotterdamthe Netherlands
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| | - Jin Li
- Department of OncologyShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Johan M Kros
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| |
Collapse
|
40
|
Wang J, Li Q, Li X, Yuan W, Huang S, Cai S, Xu J. A novel FAPα-based Z-Gly-Pro epirubicin prodrug for improving tumor-targeting chemotherapy. Eur J Pharmacol 2017; 815:166-172. [PMID: 28919026 DOI: 10.1016/j.ejphar.2017.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/09/2017] [Accepted: 09/12/2017] [Indexed: 01/28/2023]
Abstract
Fibroblast activation protein-α (FAPα) is a serine protease of the post-prolyl peptidase family that is specifically expressed in the majority of human epithelial tumors, but not in normal tissues. In this study, we demonstrated the anti-tumor activity of a novel targeting drug formed by conjugating epirubicin (EPI) with an FAPα-specific dipeptide (Z-Gly-Pro) and named it Z-GP-EPI. Consistent with this tumor-targeting delivery strategy, the results illustrated that Z-GP-EPI could release EPI efficiently after incubating with FAPα and could exhibit similar antitumor effects as EPI in vitro in FAPα over-expressed tumor cells (4T1/FAPα+). Furthermore, the evaluation of antitumor activity of Z-GP-EPI in vivo was implemented in a 4T1/FAPα+ tumor-bearing mice xenograft model. Our results illustrated that Z-GP-EPI had similar antitumor effects in 4T1/FAPα+ tumor-bearing mice and showed no visible cardiotoxicity side effects compared with free EPI. Thus, our study indicated that this FAPα-activated prodrug targeting strategy may provide a new mechanism for the targeted delivery of antitumor agents and improve their safety levels.
Collapse
Affiliation(s)
- Jun Wang
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Qiuwen Li
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Xiaojuan Li
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Weiqi Yuan
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Sichao Huang
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Shaohui Cai
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China.
| | - Jun Xu
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
41
|
Mahmoudi M, Yu M, Serpooshan V, Wu JC, Langer R, Lee RT, Karp JM, Farokhzad OC. Multiscale technologies for treatment of ischemic cardiomyopathy. NATURE NANOTECHNOLOGY 2017; 12:845-855. [PMID: 28875984 PMCID: PMC5717755 DOI: 10.1038/nnano.2017.167] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/13/2017] [Indexed: 05/02/2023]
Abstract
The adult mammalian heart possesses only limited capacity for innate regeneration and the response to severe injury is dominated by the formation of scar tissue. Current therapy to replace damaged cardiac tissue is limited to cardiac transplantation and thus many patients suffer progressive decay in the heart's pumping capacity to the point of heart failure. Nanostructured systems have the potential to revolutionize both preventive and therapeutic approaches for treating cardiovascular disease. Here, we outline recent advancements in nanotechnology that could be exploited to overcome the major obstacles in the prevention of and therapy for heart disease. We also discuss emerging trends in nanotechnology affecting the cardiovascular field that may offer new hope for patients suffering massive heart attacks.
Collapse
Affiliation(s)
- Morteza Mahmoudi
- Center for Nanomedicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| | - Mikyung Yu
- Center for Nanomedicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Vahid Serpooshan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California 94305, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Robert Langer
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Department of Medicine, Division of Cardiology, Brigham and Women’s Hospital and Harvard Medical School, Cambridge, Massachusetts 02138, USA
| | - Jeffrey M. Karp
- Center for Nanomedicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts 02139, USA
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| | - Omid C. Farokhzad
- Center for Nanomedicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| |
Collapse
|
42
|
Hanafi-Bojd MY, Moosavian Kalat SA, Taghdisi SM, Ansari L, Abnous K, Malaekeh-Nikouei B. MUC1 aptamer-conjugated mesoporous silica nanoparticles effectively target breast cancer cells. Drug Dev Ind Pharm 2017; 44:13-18. [PMID: 28832225 DOI: 10.1080/03639045.2017.1371734] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the present study, we developed aptamer (Apt) conjugated mesoporous silica nanoparticles (MSNs) for specific delivery of epirubicin (EPI) to breast cancer cells. MSNs were synthesized and functionalized with 3-mercaptopropyltrimethoxysilane (3-MPTMS), followed by MUC1 aptamer conjugation through disulfide bonds. The nanoparticles were analyzed by transmission electron microscopy (TEM), particle size analyzer, zeta potential, elemental analysis (CHNS), aptamer conjugation efficiency, drug loading efficiency, and drug release profile. Cell uptake and in vitro cytotoxicity of different formulations were performed. The results of MSNs characterization confirmed spherical nanoparticles with thiol functional groups. Particle size of obtained nanoparticles was 163 nm in deionized water. After conjugation of MUC1 aptamer and EPI loading (MSN-MUC1-EPI), particle size increased to 258 nm. The aptamer conjugation to MSNs with disulfide bonds were confirmed using gel retardation assay. Cellular uptake studies revealed better cell uptake of MSN-MUC1-EPI compared to MSN-EPI. Moreover, cytotoxicity study results in MCF7 cell lines showed improved cytotoxicity of MSN-MUC1-EPI in comparison with MSN-EPI or EPI at the same concentration of drug. These results exhibited that MSN-MUC1-EPI has the potential for targeted drug delivery into MUC1 positive breast cancer cells to improve drug efficacy and alleviate side effects.
Collapse
Affiliation(s)
- Mohammad Yahya Hanafi-Bojd
- a Cellular and Molecular Research Center, Department of Pharmacology, School of Medicine , Birjand University of Medical Sciences , Birjand , Iran
| | | | | | - Legha Ansari
- d School of Pharmacy , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Khalil Abnous
- e Pharmaceutical Research Center , Mashhad University of Medical Sciences , Mashhad , Iran
| | | |
Collapse
|
43
|
Kanchi MM, Shanmugam MK, Rane G, Sethi G, Kumar AP. Tocotrienols: the unsaturated sidekick shifting new paradigms in vitamin E therapeutics. Drug Discov Today 2017; 22:1765-1781. [PMID: 28789906 DOI: 10.1016/j.drudis.2017.08.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/01/2017] [Accepted: 08/01/2017] [Indexed: 11/15/2022]
Abstract
Vitamin E family members: tocotrienols and tocopherols are widely known for their health benefits. Decades of research on tocotrienols have shown they have diverse biological activities such as antioxidant, anti-inflammatory, anticancer, neuroprotective and skin protection benefits, as well as improved cognition, bone health, longevity and reduction of cholesterol levels in plasma. Tocotrienols also modulate several intracellular molecular targets and, most importantly, have been shown to improve lipid profiles, reduce total cholesterol and reduce the volume of white matter lesions in human clinical trials. This review provides a comprehensive update on the little-known therapeutic potentials of tocotrienols, which tocopherols lack in a variety of inflammation-driven diseases.
Collapse
Affiliation(s)
- Madhu M Kanchi
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Grishma Rane
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Alan P Kumar
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; National University Cancer Institute, National University Health System, 119074, Singapore; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia; Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
44
|
Zhou S, Zhang T, Peng B, Luo X, Liu X, Hu L, Liu Y, Di D, Song Y, Deng Y. Targeted delivery of epirubicin to tumor-associated macrophages by sialic acid-cholesterol conjugate modified liposomes with improved antitumor activity. Int J Pharm 2017; 523:203-216. [PMID: 28336455 DOI: 10.1016/j.ijpharm.2017.03.034] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/28/2017] [Accepted: 03/18/2017] [Indexed: 12/15/2022]
Abstract
With the knowledge that the receptors of sialic acid are overexpressed on the surface of tumor-associated macrophages (TAMs), which play a crucial role in the tumor's progression and metastasis, a sialic acid-cholesterol conjugate (SA-CH) was synthesized and modified on the surface of epirubicin (EPI)-loaded liposomes (EPI-SAL) to improve the delivery of EPI to the TAMs. The liposomes were developed using remote loading technology via a pH gradient. The liposomes were evaluated for particle size, encapsulation efficiency, in vitro release, stability, in vitro cytotoxicity and pharmacokinetics. And the in vitro and in vivo cellular uptake studies demonstrated EPI-SAL achieved enhanced accumulation of EPI into TAMs. The antitumor studies indicated that EPI-SAL provided the strongest antitumor activity compared with the other formulations (EPI-S, EPI-CL and EPI-PL represent EPI solution, conventional liposomal EPI, PEGylated liposomal EPI, respectively), and the survival percent of tumor-bearing mice was 83.3%. The superior antitumor efficacy was probably attributed to the killing of TAMs by EPI-SAL, and modulating the tumor microenvironment with the depletion of TAMs. These findings suggested that SA-CH decorated EPI-loaded liposomes may present an effective strategy to eradicate TAMs, which may be a promising approach for cancer therapy.
Collapse
Affiliation(s)
- Songlei Zhou
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Ting Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Bo Peng
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Xiang Luo
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Ling Hu
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Yang Liu
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Donghua Di
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, 103, Wenhua Road, Shenyang 110016, PR China, PR China.
| |
Collapse
|
45
|
Abnous K, Danesh NM, Ramezani M, Lavaee P, Jalalian SH, Yazdian-Robati R, Emrani AS, Hassanabad KY, Taghdisi SM. A novel aptamer-based DNA diamond nanostructure for in vivo targeted delivery of epirubicin to cancer cells. RSC Adv 2017. [DOI: 10.1039/c6ra28234b] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The clinical administration of epirubicin (Epi) in the treatment of cancer has been restricted, owing to its cardiotoxicity.
Collapse
Affiliation(s)
- Khalil Abnous
- Pharmaceutical Research Center
- Mashhad University of Medical Sciences
- Mashhad
- Iran
| | - Noor Mohammad Danesh
- Nanotechnology Research Center
- Mashhad University of Medical Sciences
- Mashhad
- Iran
- Research Institute of Sciences and New Technology
| | - Mohammad Ramezani
- Nanotechnology Research Center
- Mashhad University of Medical Sciences
- Mashhad
- Iran
| | - Parirokh Lavaee
- Academic Center For Education
- Culture and Research (ACECR)-Mashhad Branch
- Mashhad
- Iran
| | - Seyed Hamid Jalalian
- Nanotechnology Research Center
- Mashhad University of Medical Sciences
- Mashhad
- Iran
- Academic Center For Education
| | - Rezvan Yazdian-Robati
- Department of Pharmaceutical Biotechnology
- School of Pharmacy
- Mashhad University of Medical Sciences
- Mashhad
- Iran
| | | | - Koroush Yousefi Hassanabad
- Department of Infectious Disease
- Children Medical Center
- North Khorasan University of Medical Sciences
- Bojnord
- Iran
| | | |
Collapse
|
46
|
Yang J, Yang Q, Xu L, Lou J, Dong Z. An epirubicin-peptide conjugate with anticancer activity is dependent upon the expression level of the surface transferrin receptor. Mol Med Rep 2016; 15:323-330. [PMID: 27959443 DOI: 10.3892/mmr.2016.6004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/02/2016] [Indexed: 02/05/2023] Open
Abstract
Epirubicin (EPI) is one of the most widely used anticarcinogens; however, serious side effects, including cardiomyopathy and congestive heart failure, limit its long‑term administration. To overcome this problem, the HAIYPRH peptide ligand was used with EPI in the synthesis of a HAIYPRH‑EPI conjugate. The anticancer activity and cellular uptake of the conjugate were measured and evaluated. The results of the present study indicated that the cytotoxicity of HAIYPRH‑EPI was correlated with the expression of the cell surface transferrin receptor (TfR). The conjugate exerted high cytotoxicity and proapoptotic function when in an LN229 glioma cell line, which overexpresses surface TfR. It was hypothesized that transferrin (Tf) can promote cytotoxicity. Conversely, the conjugate exhibited very low cytotoxicity and proapoptotic function in a U87 glioma cell line, in which surface TfR expression was undetectable. In addition, fluorescence microscopy and flow cytometry methods were used to evaluate cellular uptake, and the results of these methods were consistent with the present hypotheses. The conjugate cellular uptake of the conjugate in LN229 cells was markedly higher compared with that in U87 cells, and it was hypothesized that Tf can enhance the uptake in LN229 cells. The cytotoxicity of HAIYPRH‑EPI was dependent upon the expression of surface TfR. Considering that the majority of cancer cells have high rates of iron uptake and surface TfR is generally overexpressed on cancer cells, it was speculated by the authors that HAIYPRH‑EPI may form part of an effective strategy for increasing the selectivity of EPI for cancer cells, as well as reducing its systemic toxicity. To confirm the hypothesis, the effects of HAIYPRH‑EPI on non‑cancerous cell lines were investigated. A future study will examine the side effects of HAIYPRH‑EPI, using a suitable delivery system in an animal model.
Collapse
Affiliation(s)
- Jiadan Yang
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qiyu Yang
- Department of Thoracic Oncology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lu Xu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jie Lou
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhi Dong
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
47
|
Freag MS, Elnaggar YSR, Abdelmonsif DA, Abdallah OY. Layer-by-layer-coated lyotropic liquid crystalline nanoparticles for active tumor targeting of rapamycin. Nanomedicine (Lond) 2016; 11:2975-2996. [DOI: 10.2217/nnm-2016-0236] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: This work spotlights on fabrication of CD44-tropic, layer-by-layer (LbL) coated, liquid crystalline nanoparticles of rapamycin (Rap-LbL-LCNPs) to enhance its water solubility and enable its anticancer use. Methods: Rap-LCNPs were fabricated using hydrotrope method and then coated using LbL self-assembly technique. Results: LbL coating strategy successfully reduced monoolein-induced hemolysis and increased LCNPs serum stability. Lyophilized Rap-LbL-LCNPs were successfully sterilized using γ-radiations. In CD44-overexpressed MDA-MB-231 cells, Rap-LbL-LCNPs demonstrated superior cytotoxicity compared with the nontargeted formulation. Rap-LbL-LCNPs showed 3.35-fold increase in bioavailability compared with free drug. Rap-LbL-LCNPs significantly inhibited tumor growth, enhanced animal survival and reduced nephrotoxic and hyperglycemic effects of Rap. Conclusion: LbL coating strategy of Rap-LCNPs could serve as a promising approach that facilitates Rap use in cancer therapy.
Collapse
Affiliation(s)
- May S Freag
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Yosra SR Elnaggar
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy & Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Doaa A Abdelmonsif
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
48
|
Liu MJ, Qu D, Chen Y, Liu CY, Liu YP, Ding XF. Preparation of novel butyryl galactose ester-modified coix component microemulsions and evaluation on hepatoma-targeting in vitro and in vivo. Drug Deliv 2016; 23:3444-3451. [PMID: 27198659 DOI: 10.1080/10717544.2016.1189984] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The butyryl galactose ester-modified coix component microemulsions (But-Gal-CMEs) was developed for enhanced liver tumor-specific targeting. The study was aimed to evaluate the hepatoma-targeting potential of But-Gal-CMEs in vitro and in vivo. But-Gal-CMEs with a uniform spherical shape exhibited a small particle size (56.68 ± 0.07 nm), a narrow polydispersity (PDI, 0.144 ± 0.005) and slightly negative surface charge (-0.102 ± 0.008 mV). In the cell uptake studies, But-Gal-CMEs showed a significant enhancement on the intracellular fluorescent intensity on HepG2 cells model, which was 1.93-fold higher relative to coix component microemulsions (CMEs). The IC50 of But-Gal-CMEs against HepG2 cells was 64.250 μg/mL, which was notably stronger than that of CMEs. In the cell apoptosis studies, compared with CMEs, But-Gal-CMEs (50 μg/mL) treatment resulted in a 1.34-fold rise in total apoptosis cells of HepG2. In the biodistribution studies in vivo, the intratumorous fluorescence of Cy5-loaded But-Gal-CMEs was 1.43-fold higher relative to that of Cy5-loaded CMEs, suggesting an obviously enhanced accumulation in the tumor sites. Taken as together, But-Gal could be incorporated into the coix component microemulsions as a novel ligand for realizing hepatoma-targeting drugs delivery.
Collapse
Affiliation(s)
- Ming Jian Liu
- a Multicomponent of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Provincial Academy of Chinese Medicine , Nanjing , China and.,b Department of Pharmacy , Jiangsu University , Zhen Jiang , China
| | - Ding Qu
- a Multicomponent of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Provincial Academy of Chinese Medicine , Nanjing , China and
| | - Yan Chen
- a Multicomponent of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Provincial Academy of Chinese Medicine , Nanjing , China and
| | - Cong Yan Liu
- a Multicomponent of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Provincial Academy of Chinese Medicine , Nanjing , China and
| | - Yu Ping Liu
- a Multicomponent of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Provincial Academy of Chinese Medicine , Nanjing , China and
| | - Xue Fang Ding
- a Multicomponent of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Provincial Academy of Chinese Medicine , Nanjing , China and
| |
Collapse
|
49
|
Wu J, Xue X, Zhang B, Cao H, Kong F, Jiang W, Li J, Sun D, Guo R. Enhanced antitumor activity and attenuated cardiotoxicity of Epirubicin combined with Paeonol against breast cancer. Tumour Biol 2016; 37:12301-12313. [PMID: 27272157 DOI: 10.1007/s13277-016-5088-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 05/15/2016] [Indexed: 12/13/2022] Open
Abstract
Epirubicin is widely used for the therapy of various breast cancers. However, it has serious adverse side effects, particularly cardiotoxicity, which can cause irreversible damage in patients. Paeonol, an active component from Moutan Cortex, enhances antitumor activity of antineoplastics and reduces toxicities induced by chemotherapeutics. In this study, we investigated the anticancer activity of Paeonol in combination with Epirubicin against breast cancer and the alleviated effect of Paeonol on cardiotoxicity induced by Epirubicin. The apoptosis results and the coefficient of drug interaction values suggested significantly synergistic in combination of Paeonol and Epirubicin to 4T1 and MCF-7 cells. We further examined antitumor activities of Paeonol or/and Epirubicin in vivo in BALB/c mice and found that co-treatment of Paeonol and Epirubicin had a synergistic inhibitory effect on tumor growth and enhanced apoptosis in tumors in vivo compared with Epirubicin alone. Increased apoptosis was associated with the activation of apoptosis-related proteins including PARP, Bax, caspase 3, and inhibition of p38/JNK/ERK MAPKs. Moreover, Paeonol exhibited a mitigative effect on Epirubicin-induced cardiotoxicity through suppressing NF-kB pathway. In conclusion, Paeonol (a) enhanced the antitumor activity of Epirubicin in a synergistic manner against breast cancer cells via inhibiting p38/JNK/ERK MAPKs and (b) alleviated Epirubicin-induced cardiotoxicity by suppressing NF-kB pathway. These findings suggest that combination of Paeonol and Epirubicin is potentially applicable for breast cancer treatment.
Collapse
Affiliation(s)
- Jing Wu
- Department of Pharmacy, The Second Hospital of Shandong University, 247# Beiyuan Road, Jinan, 250033, China.,Department of Pharmacology, School of Medicine, Shandong University, 44# West Wenhua Road, Jinan, 250012, China
| | - Xia Xue
- Department of Pharmacy, The Second Hospital of Shandong University, 247# Beiyuan Road, Jinan, 250033, China
| | - Bin Zhang
- Department of Pharmacy, The Second Hospital of Shandong University, 247# Beiyuan Road, Jinan, 250033, China
| | - Hongmei Cao
- Department of Pharmacy, People's Hospital of Zhangqiu, 308# Huiquan Road, Jinan, 250033, China
| | - Feng Kong
- Central Laboratory, The Second Hospital of Shandong University, 247# Beiyuan Road, Jinan, 250033, China
| | - Wen Jiang
- Central Laboratory, The Second Hospital of Shandong University, 247# Beiyuan Road, Jinan, 250033, China
| | - Juan Li
- Department of Pharmacy, The Second Hospital of Shandong University, 247# Beiyuan Road, Jinan, 250033, China
| | - Deqing Sun
- Department of Pharmacy, The Second Hospital of Shandong University, 247# Beiyuan Road, Jinan, 250033, China.
| | - Ruichen Guo
- Institute of Clinical Pharmacology, Qi Lu Hospital of Shandong University, 107# West Wenhua Road, Jinan, 250012, China.
| |
Collapse
|
50
|
Zhang L, Li G, Gao M, Liu X, Ji B, Hua R, Zhou Y, Yang Y. RGD-peptide conjugated inulin-ibuprofen nanoparticles for targeted delivery of Epirubicin. Colloids Surf B Biointerfaces 2016; 144:81-89. [PMID: 27070055 DOI: 10.1016/j.colsurfb.2016.03.077] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/16/2016] [Accepted: 03/26/2016] [Indexed: 10/22/2022]
Abstract
Recently, chemotherapy-based polymeric nanoparticles have been extensively investigated for solid tumor treatment. Tumor targeted nanoparticles demonstrated great potential for improved accumulation in the tumor tissue, superior anticancer activity and reduced side effects. Thus, inulin-ibuprofen polymer was synthesized by esterification between inulin and ibuprofen, and RGD targeted epirubicin (EPB) loaded nanoparticles were prepared by the self-assembly of inulin-ibuprofen polymer and in situ encapsulation of EPB. RGD conjugated EPB loaded nanoparticles were characterized by dynamic light scattering (DLS) and transmission electron microscope (TEM). The EPB release from the nanoparticles showed pH-dependent profile and accelerated by the decreased pH value, which would favor the effective drug delivery in vivo. Intracellular uptake analysis suggested that RGD conjugated nanoparticles could be easily internalized by the cancer cells. In vitro cytotoxicity revealed that RGD conjugated EPB loaded nanoparticles exhibited the better antitumor efficacy compared with non-conjugated nanoparticles. More importantly, RGD conjugated EPB loaded nanoparticles showed superior anticancer effects and reduced toxicity than free EPB and non-conjugated nanoparticles by in vivo antitumor activity, EPB biodistribution and histology analysis.
Collapse
Affiliation(s)
- Luzhong Zhang
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, China; Hand Surgery Research Center, Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Guicai Li
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Ming Gao
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xin Liu
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Bing Ji
- Medical College, Nantong University, Nantong 226001, China
| | - Ruheng Hua
- Medical College, Nantong University, Nantong 226001, China
| | - Youlang Zhou
- Hand Surgery Research Center, Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China.
| | - Yumin Yang
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|