1
|
Ohya Y, Ogiso Y, Matsuda M, Sakae H, Nishida K, Miki Y, Fox TE, Kester M, Sakamoto W, Nabe T, Kitatani K. Pronecroptotic Therapy Using Ceramide Nanoliposomes Is Effective for Triple-Negative Breast Cancer Cells. Cells 2024; 13:405. [PMID: 38474369 DOI: 10.3390/cells13050405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Regulated necrosis, termed necroptosis, represents a potential therapeutic target for refractory cancer. Ceramide nanoliposomes (CNLs), considered potential chemotherapeutic agents, induce necroptosis by targeting the activating protein mixed lineage kinase domain-like protein (MLKL). In the present study, we examined the potential of pronecroptotic therapy using CNLs for refractory triple-negative breast cancer (TNBC), for which there is a lack of definite and effective therapeutic targets among the various immunohistological subtypes of breast cancer. MLKL mRNA expression in tumor tissues was significantly higher in TNBC patients than in those with non-TNBC subtypes. Similarly, among the 50 breast cancer cell lines examined, MLKL expression was higher in TNBC-classified cell lines. TNBC cell lines were more susceptible to the therapeutic effects of CNLs than the non-TNBC subtypes of breast cancer cell lines. In TNBC-classified MDA-MB-231 cells, the knockdown of MLKL suppressed cell death induced by CNLs or the active substance short-chain C6-ceramide. Accordingly, TNBC cells were prone to CNL-evoked necroptotic cell death. These results will contribute to the development of CNL-based pronecroptotic therapy for TNBC.
Collapse
Affiliation(s)
- Yuki Ohya
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata 573-0101, Japan
| | - Yuri Ogiso
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata 573-0101, Japan
| | - Masaya Matsuda
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata 573-0101, Japan
| | - Harumi Sakae
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata 573-0101, Japan
| | - Kentaro Nishida
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata 573-0101, Japan
| | - Yasuhiro Miki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Todd E Fox
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908-8735, USA
| | - Mark Kester
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908-8735, USA
| | - Wataru Sakamoto
- Research Center of Oncology, Ono Pharmaceutical, Co., Ltd., Osaka 618-8585, Japan
| | - Takeshi Nabe
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata 573-0101, Japan
| | - Kazuyuki Kitatani
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata 573-0101, Japan
| |
Collapse
|
2
|
Peng Z, Ding Y, Zhang P, Lv X, Li Z, Zhou X, Huang S. Artificial Intelligence Application for Anti-tumor Drug Synergy Prediction. Curr Med Chem 2024; 31:6572-6585. [PMID: 39420717 DOI: 10.2174/0109298673290777240301071513] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/25/2024] [Accepted: 02/15/2024] [Indexed: 10/19/2024]
Abstract
Currently, the main therapeutic methods for cancer include surgery, radiation therapy, and chemotherapy. However, chemotherapy still plays an important role in tumor therapy. Due to the variety of pathogenic factors, the development process of tumors is complex and regulated by many factors, and the treatment of a single drug is easy to cause the human body to produce a drug-resistant phenotype to specific drugs and eventually leads to treatment failure. In the process of clinical tumor treatment, the combination of multiple drugs can produce stronger anti-tumor effects by regulating multiple mechanisms and can reduce the problem of tumor drug resistance while reducing the toxic side effects of drugs. Therefore, it is still a great challenge to construct an efficient and accurate screening method that can systematically consider the synergistic anti- tumor effects of multiple drugs. However, anti-tumor drug synergy prediction is of importance in improving cancer treatment outcomes. However, identifying effective drug combinations remains a complex and challenging task. This review provides a comprehensive overview of cancer drug synergy therapy and the application of artificial intelligence (AI) techniques in cancer drug synergy prediction. In addition, we discuss the challenges and perspectives associated with deep learning approaches. In conclusion, the review of the AI techniques' application in cancer drug synergy prediction can further advance our understanding of cancer drug synergy and provide more effective treatment plans and reasonable drug use strategies for clinical guidance.
Collapse
Affiliation(s)
- Zheng Peng
- Department of Clinical Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, Guangxi, China
| | - Yanling Ding
- Department of Clinical Laboratory, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Pengfei Zhang
- Department of Pulmonary and Critical Care Medicine, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, Guangxi, China
| | - Xiaolan Lv
- Department of Clinical Laboratory, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Zepeng Li
- Department of Infectious Disease, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, Guangxi, China
| | - Xiaoling Zhou
- Department of Gastroenterology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, Guangxi, China
| | - Shigao Huang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
3
|
Meng D, Ren M, Li M, Wang M, Geng W, Shang Q. Molecular mechanism of α-Hederin in tumor progression. Biomed Pharmacother 2024; 170:116097. [PMID: 38160624 DOI: 10.1016/j.biopha.2023.116097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
α-Hederin is a monosaccharide pentacyclic triterpene saponin compound derived from the Chinese herb, Pulsatilla. It has garnered considerable attention for its anti-tumor, anti-inflammatory, and spasmolytic pharmacological activities. Given the rising incidence of cancer and the pronounced adverse reactions associated with chemotherapy drugs-which profoundly impact the quality of life for cancer patients-there is an immediate need for safe and effective antitumor agents. Traditional drugs and their anticancer effects have become a focal point of research in recent years. Studies indicate that α-Hederin can hinder tumor cell proliferation and impede the advancement of various cancers, including breast, lung, colorectal, and liver cancers. The principal mechanism behind its anti-tumor activity involves inhibiting tumor cell proliferation, facilitating tumor cell apoptosis, and arresting the cell cycle process. Current evidence suggests that α-Hederin can exert its anti-tumor properties through diverse mechanisms, positioning it as a promising agent in anti-tumor therapy. However, a comprehensive literature search revealed a gap in the comprehensive understanding of α-Hederin. This paper aims to review the available literature on the anti-tumor mechanisms of α-Hederin, hoping to provide valuable insights for the clinical treatment of malignant tumors and the innovation of novel anti-tumor medications.
Collapse
Affiliation(s)
- Dandan Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Meng Ren
- Department of Physical Education, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Maofeng Li
- College of Foreign Chinese, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Min Wang
- Experimental Center of Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Wei Geng
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238, Jingshi East Road, Lixia District, Jinan 250014, China
| | - Qingxin Shang
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China.
| |
Collapse
|
4
|
Sandal P, Kumari L, Patel P, Singh A, Singh D, Gupta GD, Kurmi BD. Doxorubicin Conjugates: An Efficient Approach for Enhanced Therapeutic Efficacy with Reduced Side Effects. Assay Drug Dev Technol 2023; 21:137-156. [PMID: 37083490 DOI: 10.1089/adt.2022.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Continuous drug delivery modification is the scientific approach and is a basic need for the efficient therapeutic efficacy of active drug molecules. Polymer-drug conjugates have long been a hallmark of the drug delivery sector, with various conjugates on the market or in clinical trials. Improved drug solubilization, extended blood circulation, decreased immunogenicity, controlled release behavior, and increased safety are the advantages of conjugating drugs to the polymeric carrier like polyethylene glycol (PEG). Polymer therapies have evolved over the last decade, resulting in polymer-drug conjugates with diverse topologies and chemical properties. Traditional nondegradable polymeric carriers like PEG and hydroxy propyl methacrylate have been clinically employed to fabricate polymer-drug conjugates. Still, functionalized polymer-drug conjugates are increasingly being used to increase localized drug delivery and ease of removal. Researchers have developed multifunctional carriers that can "see and treat" patients using medicinal and diagnostic chemicals. This review focused on the various conjugation approaches for attaching the doxorubicin to different polymers to achieve enhanced therapeutic efficacy, that is, increased bioavailability and reduced adverse effects.
Collapse
Affiliation(s)
- Pallavi Sandal
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Lakshmi Kumari
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Punjab, India
| | - Amrinder Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Dilpreet Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | | | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| |
Collapse
|
5
|
Zhang X, Zhu L, Wang X, Zhang H, Wang L, Xia L. Basic research on curcumin in cervical cancer: Progress and perspectives. Biomed Pharmacother 2023; 162:114590. [PMID: 36965256 DOI: 10.1016/j.biopha.2023.114590] [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: 12/31/2022] [Revised: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023] Open
Abstract
Curcumin is a polyphenolic substance extracted from plants such as Curcuma longa, Curcuma zedoaria, and radix curcumae, and it has attracted much attention because of the anti-inflammatory, antioxidant, anti-tumor, antibacterial and other multiple pharmacological effects. Cervical cancer is one of the most common malignant tumors in women. With the application of HPV (human papillomavirus) vaccine, the incidence of cervical cancer is expected to be reduced, but it remains difficult to promote the vaccine among low-income population. As a commonly used food additive, curcumin has recently been found to have a significant therapeutic effect in the treatment of cervical cancer. In recent years, numerous in vitro and in vivo studies have found that curcumin can have significant efficacy in anti-cervical cancer treatment by promoting apoptosis, inhibiting tumour cell proliferation, metastasis and invasion, inhibiting HPV and inducing autophagy in tumour cells. However, due to poor water solubility, rapid catabolism, and low bioavailability of curcumin, studies on curcumin derivatives and novel formulations are increasing. Curcumin has a wide range of mechanisms of action against cervical cancer and may become a novel antitumor drug in the future, opening up new ideas for the research of curcumin in the field of antitumor. There is a lack of systematic reviews on the mechanism of action of curcumin against cervical cancer. Therefore, this study is a review of the literature based on the mechanism of action of curcumin against cervical cancer, with a view to providing reference information for scientific and clinical practitioners.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Zhu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuezhen Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hairong Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Jinan, China
| | - Lianzhong Wang
- Department of Respiratory and Critical Care Medicine of Second affiliated hospital, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Lei Xia
- Department of Pathology, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
6
|
Chen J, Yu X, Liu X, Ni J, Yang G, Zhang K. Advances in nanobiotechnology-propelled multidrug resistance circumvention of cancer. NANOSCALE 2022; 14:12984-12998. [PMID: 36056710 DOI: 10.1039/d2nr04418h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Multidrug resistance (MDR) is one of the main reasons for the failure of tumor chemotherapy and has a negative influence on the therapeutic effect. MDR is primarily attributable to two mechanisms: the activation of efflux pumps for drugs, which can transport intracellular drug molecules from cells, and other mechanisms not related to efflux pumps, e.g., apoptosis prevention, strengthened DNA repair, and strong oxidation resistance. Nanodrug-delivery systems have recently attracted much attention, showing some unparalleled advantages such as drug targeting and reduced drug efflux, drug toxicity and side effects in reversing MDR. Notably, in drug-delivery platforms based on nanotechnology, multiple therapeutic strategies are integrated into one system, which can compensate for the limitations of individual strategies. In this review, the mechanisms of tumor MDR as well as common vectors and nanocarrier-combined therapy strategies to reverse MDR were summarized to promote the understanding of the latest progress in improving the efficiency of chemotherapy and synergistic strategies. In particular, the adoption of nanotechnology has been highlighted and the principles underlying this phenomenon have been elucidated, which may provide guidance for the development of more effective anticancer strategies.
Collapse
Affiliation(s)
- Jie Chen
- Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China.
- Department of Medical Ultrasound, Shanghai Chest Hospital, Shanghai Jiao Tong University, No.241 West Huaihai Road, Shanghai 200030, P. R. China
| | - Xin Yu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Xinyu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, No. 507 Zheng-Min Road, Shanghai 200433, P. R. China
| | - Jinliang Ni
- Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China.
| | - Guangcan Yang
- Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China.
| | - Kun Zhang
- Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, P. R. China.
| |
Collapse
|
7
|
Zhang M, Zhang Z, Song X, Zhu J, Sng JA, Li J, Wen Y. Synthesis and Characterization of Palmitoyl- block-poly(methacryloyloxyethyl Phosphorylcholine) Polymer Micelles for Anticancer Drug Delivery. Biomacromolecules 2022; 23:4586-4596. [DOI: 10.1021/acs.biomac.2c00838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Miao Zhang
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Zhongxing Zhang
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Xia Song
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Jing An Sng
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jun Li
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
- National University of Singapore (Chongqing) Research Institute, 2 Huizhu Road, Yubei District, Chongqing 401120, China
| | - Yuting Wen
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- National University of Singapore (Chongqing) Research Institute, 2 Huizhu Road, Yubei District, Chongqing 401120, China
| |
Collapse
|
8
|
Emran TB, Shahriar A, Mahmud AR, Rahman T, Abir MH, Siddiquee MFR, Ahmed H, Rahman N, Nainu F, Wahyudin E, Mitra S, Dhama K, Habiballah MM, Haque S, Islam A, Hassan MM. Multidrug Resistance in Cancer: Understanding Molecular Mechanisms, Immunoprevention and Therapeutic Approaches. Front Oncol 2022; 12:891652. [PMID: 35814435 PMCID: PMC9262248 DOI: 10.3389/fonc.2022.891652] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide. Several treatments are available for cancer treatment, but many treatment methods are ineffective against multidrug-resistant cancer. Multidrug resistance (MDR) represents a major obstacle to effective therapeutic interventions against cancer. This review describes the known MDR mechanisms in cancer cells and discusses ongoing laboratory approaches and novel therapeutic strategies that aim to inhibit, circumvent, or reverse MDR development in various cancer types. In this review, we discuss both intrinsic and acquired drug resistance, in addition to highlighting hypoxia- and autophagy-mediated drug resistance mechanisms. Several factors, including individual genetic differences, such as mutations, altered epigenetics, enhanced drug efflux, cell death inhibition, and various other molecular and cellular mechanisms, are responsible for the development of resistance against anticancer agents. Drug resistance can also depend on cellular autophagic and hypoxic status. The expression of drug-resistant genes and the regulatory mechanisms that determine drug resistance are also discussed. Methods to circumvent MDR, including immunoprevention, the use of microparticles and nanomedicine might result in better strategies for fighting cancer.
Collapse
Affiliation(s)
- Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Asif Shahriar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, United States
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| | | | - Hossain Ahmed
- Department of Biotechnology and Genetic Engineering, University of Development Alternative, Dhaka, Bangladesh
| | - Nova Rahman
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Dhaka, Bangladesh
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Elly Wahyudin
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Mahmoud M Habiballah
- Medical Laboratory Technology Department, Jazan University, Jazan, Saudi Arabia
- SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Bursa Uludağ University Faculty of Medicine, Bursa, Turkey
| | | | - Mohammad Mahmudul Hassan
- Queensland Alliance for One Health Sciences, School of Veterinary Science, The University of Queensland, Gatton, QLD, Australia
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| |
Collapse
|
9
|
Targeting lipid metabolism in the treatment of ovarian cancer. Oncotarget 2022; 13:768-783. [PMID: 35634242 PMCID: PMC9132258 DOI: 10.18632/oncotarget.28241] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 05/07/2022] [Indexed: 11/25/2022] Open
Abstract
Cancer cells undergo alterations in lipid metabolism to support their high energy needs, tumorigenesis and evade an anti-tumor immune response. Alterations in fatty acid production are controlled by multiple enzymes, chiefly Acetyl CoA Carboxylase, ATP-Citrate Lyase, Fatty Acid Synthase, and Stearoyl CoA Desaturase 1. Ovarian cancer (OC) is a common gynecological malignancy with a high rate of aggressive carcinoma progression and drug resistance. The accumulation of unsaturated fatty acids in ovarian cancer supports cell growth, increased cancer cell migration, and worse patient outcomes. Ovarian cancer cells also expand their lipid stores via increased uptake of lipids using fatty acid translocases, fatty acid-binding proteins, and low-density lipoprotein receptors. Furthermore, increased lipogenesis and lipid uptake promote chemotherapy resistance and dampen the adaptive immune response needed to eliminate tumors. In this review, we discuss the role of lipid synthesis and metabolism in driving tumorigenesis and drug resistance in ovarian cancer conferring poor prognosis and outcomes in patients. We also cover some aspects of how lipids fuel ovarian cancer stem cells, and how these metabolic alterations in intracellular lipid content could potentially serve as biomarkers of ovarian cancer.
Collapse
|
10
|
Development of liposomal formulations of the eggplant glycoalkaloids solasonine and solamargine. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Kim M, Park SC, Lee DY. Glycyrrhizin as a Nitric Oxide Regulator in Cancer Chemotherapy. Cancers (Basel) 2021; 13:cancers13225762. [PMID: 34830916 PMCID: PMC8616433 DOI: 10.3390/cancers13225762] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Glycyrrhizin (GL) has anti-cancer, anti-inflammatory, anti-viral, and anti-oxidant activity. In particular, GL reduces multidrug resistance (MDR) in cancer cells, which is a major obstacle to chemotherapy. Nitric oxide (NO) also plays an important role in MDR, and GL affects NO concentration in the tumor microenvironment. However, the effects of GL and NO interaction on MDR have not been reviewed. Here, we review the role of GL as an NO regulator in cancer cells and its subsequent anti-MDR effect and posit that GL is a promising MDR inhibitor for cancer chemotherapy. Abstract Chemotherapy is used widely for cancer treatment; however, the evolution of multidrug resistance (MDR) in many patients limits the therapeutic benefits of chemotherapy. It is important to overcome MDR for enhanced chemotherapy. ATP-dependent efflux of drugs out of cells is the main mechanism of MDR. Recent studies have suggested that nitric oxide (NO) can be used to overcome MDR by inhibiting the ATPase function of ATP-dependent pumps. Several attempts have been made to deliver NO to the tumor microenvironment (TME), however there are limitations in delivery. Glycyrrhizin (GL), an active compound of licorice, has been reported to both reduce the MDR effect by inhibiting ATP-dependent pumps and function as a regulator of NO production in the TME. In this review, we describe the potential role of GL as an NO regulator and MDR inhibitor that efficiently reduces the MDR effect in cancer chemotherapy.
Collapse
Affiliation(s)
- Minsu Kim
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea; (M.K.); (S.C.P.)
| | - Seok Chan Park
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea; (M.K.); (S.C.P.)
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea; (M.K.); (S.C.P.)
- Institute of Nano Science & Technology (INST), Hanyang University, Seoul 04763, Korea
- Elixir Pharmatech Inc., Seoul 04763, Korea
- Correspondence:
| |
Collapse
|
12
|
Xia W, Tao Z, Zhu B, Zhang W, Liu C, Chen S, Song M. Targeted Delivery of Drugs and Genes Using Polymer Nanocarriers for Cancer Therapy. Int J Mol Sci 2021; 22:9118. [PMID: 34502028 PMCID: PMC8431379 DOI: 10.3390/ijms22179118] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/16/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer is one of the primary causes of worldwide human deaths. Most cancer patients receive chemotherapy and radiotherapy, but these treatments are usually only partially efficacious and lead to a variety of serious side effects. Therefore, it is necessary to develop new therapeutic strategies. The emergence of nanotechnology has had a profound impact on general clinical treatment. The application of nanotechnology has facilitated the development of nano-drug delivery systems (NDDSs) that are highly tumor selective and allow for the slow release of active anticancer drugs. In recent years, vehicles such as liposomes, dendrimers and polymer nanomaterials have been considered promising carriers for tumor-specific drug delivery, reducing toxicity and improving biocompatibility. Among them, polymer nanoparticles (NPs) are one of the most innovative methods of non-invasive drug delivery. Here, we review the application of polymer NPs in drug delivery, gene therapy, and early diagnostics for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Siyu Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (W.X.); (Z.T.); (B.Z.); (W.Z.); (C.L.)
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (W.X.); (Z.T.); (B.Z.); (W.Z.); (C.L.)
| |
Collapse
|
13
|
Bei Y, Chen X, Xu Q, Lv J, Hu J, Yang S. Apatinib weakens resistance of gastric cancer cells to paclitaxel by suppressing JAK/STAT3 signaling pathway. Drug Dev Res 2021; 83:379-388. [PMID: 34405891 DOI: 10.1002/ddr.21867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 01/16/2023]
Abstract
Apatinib has experienced a long-term study in enhancing the sensitivity of various cancer cells to chemotherapy drugs. Currently, researches show that apatinib could attenuate the resistance of gastric cancer (GC) cells to paclitaxel (PTX), but the mechanism has not been fully elucidated, which therefore was explored in this study. PTX-resistant GC cell, namely HGC-27R, was established by exposure to stepwise-increasing PTX. The cell viability of HGC-27 and HGC-27R under PTX or apatinib at different concentrations was assessed by CCK-8 assay, while scratching test and invasion assay were used for investigating the harmful influence of GC cells resistance to PTX. The function of apatinib in HGC-27R was studied by performing functional experiments (flow cytometry, scratching test, and invasion assay). Western blot was performed to measure the expressions of proteins concerning apoptosis, epithelial-mesenchymal transition and janus-activated kinase (JAK)/signal transducer and activator of transcription (STAT) pathway. PTX-resistant GC cell, namely HGC-27R, was successively constructed. HGC-27R cells showed resistance to PTX by promoting migratory and invasive capabilities. Apatinib not only inhibited cell viability of HGC-27 and HGC-27R, but also combined with PTX to suppress that of HGC-27R. Apatinib enhanced apoptosis, diminished migration and invasion of HGC-27R cells, elevated proapoptotic protein expression, and reduced Bcl-2, vimentin, snail, MMP-3, MMP-2, and MMP-9 expressions. The phosphorylation of JAK2 and STAT3 was repressed by apatinib. JAK2 partially reversed the effect of apatinib on enhancing sensitivity of GC cells to PTX. Apatinib strengthened sensitivity of GC cells to PTX by inhibiting JAK/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yanping Bei
- Department of Radiotherapy, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Xue Chen
- Department of Radiotherapy, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Quan Xu
- Department of Radiotherapy, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Jiaming Lv
- Department of Radiotherapy, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Jing Hu
- Department of Radiotherapy, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Shaohui Yang
- Department of Anorectal Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| |
Collapse
|
14
|
Subhan MA, Yalamarty SSK, Filipczak N, Parveen F, Torchilin VP. Recent Advances in Tumor Targeting via EPR Effect for Cancer Treatment. J Pers Med 2021; 11:571. [PMID: 34207137 PMCID: PMC8234032 DOI: 10.3390/jpm11060571] [Citation(s) in RCA: 223] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer causes the second-highest rate of death world-wide. A major shortcoming inherent in most of anticancer drugs is their lack of tumor selectivity. Nanodrugs for cancer therapy administered intravenously escape renal clearance, are unable to penetrate through tight endothelial junctions of normal blood vessels and remain at a high level in plasma. Over time, the concentration of nanodrugs builds up in tumors due to the EPR effect, reaching several times higher than that of plasma due to the lack of lymphatic drainage. This review will address in detail the progress and prospects of tumor-targeting via EPR effect for cancer therapy.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, Shah Jalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Satya Siva Kishan Yalamarty
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; (S.S.K.Y.); (N.F.); (F.P.)
| | - Nina Filipczak
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; (S.S.K.Y.); (N.F.); (F.P.)
| | - Farzana Parveen
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; (S.S.K.Y.); (N.F.); (F.P.)
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Punjab 63100, Pakistan
| | - Vladimir P. Torchilin
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; (S.S.K.Y.); (N.F.); (F.P.)
- Department of Oncology, Radiotherapy and Plastic Surgery, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
15
|
Paus C, van der Voort R, Cambi A. Nanomedicine in cancer therapy: promises and hurdles of polymeric nanoparticles. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The limitations of current cancer treatments have stimulated the application of nanotechnology to develop more effective and safer cancer therapies. Remarkable progress has been made in the development of nanomedicine to overcome issues associated with conventional cancer treatment, including low drug solubility, insufficient targeting, and drug resistance. The modulation of nanoparticles allows the improvement of drug pharmacokinetics, leading to improved targeting and reduced side effects. In addition, nanoparticles can be conjugated to ligands that specifically target cancer cells. Furthermore, strategies that exploit tumor characteristics to locally trigger drug release have shown to increase targeted drug delivery. However, although some clinical successes have been achieved, most nanomedicines fail to reach the clinic. Factors that hinder clinical translation vary from the complexity of design, incomplete understanding of biological mechanisms, and high demands during the manufacturing process. Clinical translation might be improved by combining knowledge from different disciplines such as cell biology, chemistry, and tumor pathophysiology. An increased understanding on how nanoparticle modifications affect biological systems is pivotal to improve design, eventually aiding development of more effective nanomedicines. This review summarizes the key successes that have been made in nanomedicine, including improved drug delivery and release by polymeric nanoparticles as well as the introduction of strategies that overcome drug resistance. In addition, the application of nanomedicine in immunotherapy is discussed, and several remaining challenges addressed.
Collapse
|
16
|
Wei X, Song M, Li W, Huang J, Yang G, Wang Y. Multifunctional nanoplatforms co-delivering combinatorial dual-drug for eliminating cancer multidrug resistance. Am J Cancer Res 2021; 11:6334-6354. [PMID: 33995661 PMCID: PMC8120214 DOI: 10.7150/thno.59342] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/26/2021] [Indexed: 02/05/2023] Open
Abstract
Clinically, the primary cause of chemotherapy failure belongs to the occurrence of cancer multidrug resistance (MDR), which directly leads to the recurrence and metastasis of cancer along with high mortality. More and more attention has been paid to multifunctional nanoplatform-based dual-therapeutic combination to eliminate resistant cancers. In addition to helping both cargoes improve hydrophobicity and pharmacokinetic properties, increase bioavailability, release on demand and enhance therapeutic efficacy with low toxic effects, these smart co-delivery nanocarriers can even overcome drug resistance. Here, this review will not only present different types of co-delivery nanocarriers, but also summarize targeted and stimuli-responsive combination nanomedicines. Furthermore, we will focus on the recent progress in the co-delivery of dual-drug using such intelligent nanocarriers for surmounting cancer MDR. Whereas it remains to be seriously considered that there are some knotty issues in the fight against MDR of cancers via using co-delivery nanoplatforms, including limited intratumoral retention, the possible changes of combinatorial ratio under complex biological environments, drug release sequence from the nanocarriers, and subsequent free-drug resistance after detachment from the nanocarriers. It is hoped that, with the advantage of continuously developing nanomaterials, two personalized therapeutic agents in combination can be better exploited to achieve the goal of cooperatively combating cancer MDR, thus advancing the time to clinical transformation.
Collapse
|
17
|
Zhang X, Sakamoto W, Canals D, Ishibashi M, Matsuda M, Nishida K, Toyoshima M, Shigeta S, Taniguchi M, Senkal CE, Okazaki T, Yaegashi N, Hannun YA, Nabe T, Kitatani K. Ceramide synthase 2-C 24:1 -ceramide axis limits the metastatic potential of ovarian cancer cells. FASEB J 2021; 35:e21287. [PMID: 33423335 PMCID: PMC8237407 DOI: 10.1096/fj.202001504rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/28/2020] [Accepted: 12/03/2020] [Indexed: 12/23/2022]
Abstract
Regulation of sphingolipid metabolism plays a role in cellular homeostasis, and dysregulation of these pathways is involved in cancer progression. Previously, our reports identified ceramide as an anti-metastatic lipid. In the present study, we investigated the biochemical alterations in ceramide-centered metabolism of sphingolipids that were associated with metastatic potential. We established metastasis-prone sublines of SKOV3 ovarian cancer cells using an in vivo selection method. These cells showed decreases in ceramide levels and ceramide synthase (CerS) 2 expression. Moreover, CerS2 downregulation in ovarian cancer cells promoted metastasis in vivo and potentiated cell motility and invasiveness. Moreover, CerS2 knock-in suppressed the formation of lamellipodia required for cell motility in this cell line. In order to define specific roles of ceramide species in cell motility controlled by CerS2, the effect of exogenous long- and very long-chain ceramide species on the formation of lamellipodia was evaluated. Treatment with distinct ceramides increased cellular ceramides and had inhibitory effects on the formation of lamellipodia. Interestingly, blocking the recycling pathway of ceramides by a CerS inhibitor was ineffective in the suppression of exogenous C24:1 -ceramide for the formation of lamellipodia. These results suggested that C24:1 -ceramide, a CerS2 metabolite, predominantly suppresses the formation of lamellipodia without the requirement for deacylation/reacylation. Moreover, knockdown of neutral ceramidase suppressed the formation of lamellipodia concomitant with upregulation of C24:1 -ceramide. Collectively, the CerS2-C24:1 -ceramide axis, which may be countered by neutral ceramidase, is suggested to limit cell motility and metastatic potential. These findings may provide insights that lead to further development of ceramide-based therapy and biomarkers for metastatic ovarian cancer.
Collapse
Affiliation(s)
- Xuewei Zhang
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Wataru Sakamoto
- Department of Medicine, Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Daniel Canals
- Department of Medicine, Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Masaya Matsuda
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Science, Setsunan University, Osaka, Japan
| | - Kentaro Nishida
- Department of Integrative Pharmaceutical Sciences, Faculty of Pharmaceutical Science, Setsunan University, Osaka, Japan
| | - Masafumi Toyoshima
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Shogo Shigeta
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Makoto Taniguchi
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Can E. Senkal
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, VA, USA
| | - Toshiro Okazaki
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
- Department of Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yusuf A. Hannun
- Department of Medicine, Stony Brook Cancer Center, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry, Stony Brook University, Stony Brook, NY, USA
| | - Takeshi Nabe
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Science, Setsunan University, Osaka, Japan
| | - Kazuyuki Kitatani
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Science, Setsunan University, Osaka, Japan
| |
Collapse
|
18
|
Melvin RG, Chaudhry N, Georgewill O, Freese R, Simmons GE. Predictive power of SARS-CoV-2 wastewater surveillance for diverse populations across a large geographical range. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.23.21250376. [PMID: 33532795 PMCID: PMC7852246 DOI: 10.1101/2021.01.23.21250376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The COVID-19 pandemic has exacerbated the disparities in healthcare delivery in the US. Many communities had, and continue to have, limited access to COVID-19 testing, making it difficult to track the spread and impact of COVID-19 in early days of the outbreak. To address this issue we monitored severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA at the population-level using municipal wastewater influent from 19 cities across the state of Minnesota during the COVID-19 outbreak in Summer 2020. Viral RNA was detected in wastewater continually for 20-weeks for cities ranging in populations from 500 to >1, 000, 000. Using a novel indexing method, we were able to compare the relative levels of SARS-CoV-2 RNA for each city during this sampling period. Our data showed that viral RNA trends appeared to precede clinically confirmed cases across the state by several days. Lag analysis of statewide trends confirmed that wastewater SARS-CoV-2 RNA levels preceded new clinical cases by 15-17 days. At the regional level, new clinical cases lagged behind wastewater viral RNA anywhere from 4-20 days. Our data illustrates the advantages of monitoring at the population-level to detect outbreaks. Additionally, by tracking infections with this unbiased approach, resources can be directed to the most impacted communities before the need outpaces the capacity of local healthcare systems.
Collapse
Affiliation(s)
- Richard G. Melvin
- Department of Biomedical Sciences, University of Minnesota Medical School – Duluth, MN
| | - Nabiha Chaudhry
- Department of Biomedical Sciences, University of Minnesota Medical School – Duluth, MN
| | - Onimitein Georgewill
- National Summer Undergraduate Research Program, University of Maryland – College Park, MD
| | - Rebecca Freese
- Biostatistical Design and Analysis Center, Clinical and Translational Science Institute, University of Minnesota – Minneapolis, MN
| | - Glenn E. Simmons
- Department of Biomedical Sciences, University of Minnesota Medical School – Duluth, MN
- Carcinogenesis and Chemoprevention Program, Masonic Cancer Center – Minneapolis, MN
| |
Collapse
|
19
|
Kolluru LP, Chandran T, Shastri PN, Rizvi SAA, D’Souza MJ. Development and evaluation of polycaprolactone based docetaxel nanoparticle formulation for targeted breast cancer therapy. JOURNAL OF NANOPARTICLE RESEARCH 2020; 22:372. [DOI: 10.1007/s11051-020-05096-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/24/2020] [Indexed: 10/16/2023]
|
20
|
Das SS, Bharadwaj P, Bilal M, Barani M, Rahdar A, Taboada P, Bungau S, Kyzas GZ. Stimuli-Responsive Polymeric Nanocarriers for Drug Delivery, Imaging, and Theragnosis. Polymers (Basel) 2020; 12:E1397. [PMID: 32580366 PMCID: PMC7362228 DOI: 10.3390/polym12061397] [Citation(s) in RCA: 221] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
In the past few decades, polymeric nanocarriers have been recognized as promising tools and have gained attention from researchers for their potential to efficiently deliver bioactive compounds, including drugs, proteins, genes, nucleic acids, etc., in pharmaceutical and biomedical applications. Remarkably, these polymeric nanocarriers could be further modified as stimuli-responsive systems based on the mechanism of triggered release, i.e., response to a specific stimulus, either endogenous (pH, enzymes, temperature, redox values, hypoxia, glucose levels) or exogenous (light, magnetism, ultrasound, electrical pulses) for the effective biodistribution and controlled release of drugs or genes at specific sites. Various nanoparticles (NPs) have been functionalized and used as templates for imaging systems in the form of metallic NPs, dendrimers, polymeric NPs, quantum dots, and liposomes. The use of polymeric nanocarriers for imaging and to deliver active compounds has attracted considerable interest in various cancer therapy fields. So-called smart nanopolymer systems are built to respond to certain stimuli such as temperature, pH, light intensity and wavelength, and electrical, magnetic and ultrasonic fields. Many imaging techniques have been explored including optical imaging, magnetic resonance imaging (MRI), nuclear imaging, ultrasound, photoacoustic imaging (PAI), single photon emission computed tomography (SPECT), and positron emission tomography (PET). This review reports on the most recent developments in imaging methods by analyzing examples of smart nanopolymers that can be imaged using one or more imaging techniques. Unique features, including nontoxicity, water solubility, biocompatibility, and the presence of multiple functional groups, designate polymeric nanocues as attractive nanomedicine candidates. In this context, we summarize various classes of multifunctional, polymeric, nano-sized formulations such as liposomes, micelles, nanogels, and dendrimers.
Collapse
Affiliation(s)
- Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India;
| | - Priyanshu Bharadwaj
- UFR des Sciences de Santé, Université de Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
| | - Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 76175-133, Iran;
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Pablo Taboada
- Colloids and Polymers Physics Group, Condensed Matter Physics Area, Particle Physics Department Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - George Z. Kyzas
- Department of Chemistry, International Hellenic University, 65404 Kavala, Greece
| |
Collapse
|
21
|
Shanmugam T, Joshi N, Ahamad N, Deshmukh A, Banerjee R. Enhanced absorption, and efficacy of oral self-assembled paclitaxel nanocochleates in multi-drug resistant colon cancer. Int J Pharm 2020; 586:119482. [PMID: 32492505 DOI: 10.1016/j.ijpharm.2020.119482] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 05/15/2020] [Accepted: 05/25/2020] [Indexed: 11/18/2022]
Abstract
Chemotherapy in drug-resistant cancers remains a challenge. Owing to associated poor bioavailability, oral administration of hydrophobic anticancer drugs like paclitaxel has been quite challenging, with the scenario being further complicated by Pgp efflux in drug-resistant tumours. We developed a novel nanocochleates (CPT) system encapsulating paclitaxel (PTX) to treat resistant colon cancer by oral administration. PTX encapsulated nanocochleates (PTX-CPT), made up of phosphatidylserine in size range of 350-600 nm with -20 ± 5.2 mV zeta potential were protected from degradation at acidic gastric pH and showed sustained PTX release over 48 h under intestinal pH condition. In vitro cytotoxicity studies on HCT-116 & HCT-15 cells (multi-drug resistant) established IC50 value of <10 and 69 nM, respectively, which was significantly lower when compared to commercial Taxol formulation. Further, the in vivo efficacy with five oral doses of 30 mg/kg PTX-CPT in an HCT-15 drug-resistant colon cancer xenograft mouse model showed more than 25 fold reduction in the tumour growth inhibition as compared to intravenous Taxol which showed just 1.94% inhibition. Interestingly, PTX-CPT treated mice also showed significantly lower proliferation index and microvessel density when compared to Taxol treated mice. Nanocochleates showed lower toxicity with at LD-50 value greater than 300 mg/kg as described in OECD 423 guideline. The enhanced efficacy of PTX-CPT speculated due to its internalization by active endocytosis, ability to escape Pgp efflux, and due to a combined effect of the pro-apoptotic and antiangiogenic role. Taken together, the results suggested the PTX-CPT a promising strategy for efficiently treating drug-resistant colon cancer orally.
Collapse
Affiliation(s)
- Thanigaivel Shanmugam
- Department of Biosciences and Bioengineering, Indian Institute of Technology-Bombay, Mumbai 400076, India
| | - Nitin Joshi
- Department of Biosciences and Bioengineering, Indian Institute of Technology-Bombay, Mumbai 400076, India
| | - Nadim Ahamad
- Department of Biosciences and Bioengineering, Indian Institute of Technology-Bombay, Mumbai 400076, India
| | - Atul Deshmukh
- Oral & Maxillofacial Pathology & Immunohistochemistry Centre, Mumbai 400003, India
| | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology-Bombay, Mumbai 400076, India.
| |
Collapse
|
22
|
Choi MK, Song IS. Recent advances in the formulation of sphingolipid anticancer therapeutics. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2020. [DOI: 10.1007/s40005-020-00475-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
23
|
PKCζ mediated anti-proliferative effect of C2 ceramide on neutralization of the tumor microenvironment and melanoma regression. Cancer Immunol Immunother 2020; 69:611-627. [PMID: 31996991 DOI: 10.1007/s00262-020-02492-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 01/18/2020] [Indexed: 12/12/2022]
Abstract
Immunotherapy, which has advantages over chemotherapy due to lesser toxicity and higher specificity, is on the rise to treat cancer. Recently, pro-apoptotic glycolipid, ceramide has emerged as a key regulator in cancer immunotherapy. The present study elucidated the potential anti-melanoma efficacy of cell-permeable, exogenous C2 ceramide on cell death and amelioration of tumor microenvironment (TME). We, for the first time, demonstrated that C2 ceramide triggered apoptosis of melanoma cells by augmenting PKCζ along with pro-inflammatory cytokines and signaling factors. C2 ceramide showed a PKCζ-mediated tumor-suppressive role in melanoma without exhibiting hepatotoxicity and nephrotoxicity. Moreover, PKCζ was revealed as one of the key regulators of Akt and ceramide during C2 ceramide-mediated apoptosis. C2 ceramide was effective in repolarization of M2 macrophage phenotype and reduction of angiogenic factors such as VEGF, VEGFR1, VEGFR2, HIF1α. Interestingly, PKCζ knockdown attenuated C2 ceramide-mediated inhibition of melanoma progression. Restoration of the Th1 type TME by C2 ceramide enhanced cytotoxic T cell-mediated killing of melanoma cells. Altogether, the study unraveled that C2 ceramide-induced PKCζ was associated with favorable immune cell functioning in TME leading to melanoma regression. Thus, our findings explored a novel mechanistic insight into C2 ceramide as a promising immunotherapeutic agent in melanoma treatment.
Collapse
|
24
|
Chen J, Khiste SK, Fu X, Roy KR, Dong Y, Zhang J, Liu M, Liu YY, Liu Z. Rubusoside-assisted solubilization of poorly soluble C 6-Ceramide for a pilot pharmacokinetic study. Prostaglandins Other Lipid Mediat 2019; 146:106402. [PMID: 31841664 DOI: 10.1016/j.prostaglandins.2019.106402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/22/2019] [Accepted: 12/03/2019] [Indexed: 12/30/2022]
Abstract
Although C6-Ceramide has attracted much attention as a possible tumor suppressor, the delivery of C6-Ceramide is still challenging due to its inherent hydrophobicity and insolubility. In this study we explored the use of a natural compound rubusoside (RUB) as a solubilizer to enhance the solubility of a fluorescence-labeled C6-Ceramide (NBD C6-Ceramide) and to characterize its pharmacokinetics and tissue distribution in an animal model. RUB significantly enhanced the solubility of NBD C6-Ceramide by forming nanomicelles, and efficiently delivered NBD C6-Ceramide in rats by oral and intravenous administration. RUB loaded 1.96 % of NBD C6-Ceramide in the nanomicelles and solubilized it to a concentration of 3.6 mg/mL in water. NBD C6-Ceramide in nanomicelles remained stable in aqueous solutions, allowing intravenous administration without the use of any organic solvents or surfactants. After oral administration, NBD C6-Ceramide rapidly rose to peak plasma concentrations within the first 90 min, distributed to tissues, and remained in vivo for more than 24 h. Tissular levels of NBD C6-Ceramide from high to low were associated with heart, lung, cerebellum, testicle, spleen, liver, kidney, and brain. Altogether, our study demonstrated that RUB-assisted nanomicelles can serve as an efficient and convenient delivery system for short-chain C6-Ceramide and enable in vivo evaluation of potential new cancer treatments.
Collapse
Affiliation(s)
- Jianzhong Chen
- School of Renewable Natural Resources, LSU Agricultural Center, Louisiana State University, Baton Rouge 70803, USA
| | - Sachin K Khiste
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71203, USA
| | - Xiaomei Fu
- School of Renewable Natural Resources, LSU Agricultural Center, Louisiana State University, Baton Rouge 70803, USA; School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Kartik R Roy
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71203, USA
| | - Yixuan Dong
- School of Renewable Natural Resources, LSU Agricultural Center, Louisiana State University, Baton Rouge 70803, USA
| | - Jian Zhang
- School of Renewable Natural Resources, LSU Agricultural Center, Louisiana State University, Baton Rouge 70803, USA; School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Mei Liu
- School of Renewable Natural Resources, LSU Agricultural Center, Louisiana State University, Baton Rouge 70803, USA; School of Pharmacy, Guangzhou University of Traditional Chinese Medicine, Guangzhou 330004, China
| | - Yong-Yu Liu
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71203, USA
| | - Zhijun Liu
- School of Renewable Natural Resources, LSU Agricultural Center, Louisiana State University, Baton Rouge 70803, USA.
| |
Collapse
|
25
|
Bidram E, Esmaeili Y, Ranji-Burachaloo H, Al-Zaubai N, Zarrabi A, Stewart A, Dunstan DE. A concise review on cancer treatment methods and delivery systems. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101350] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
26
|
Khiste SK, Liu Z, Roy KR, Uddin MB, Hosain SB, Gu X, Nazzal S, Hill RA, Liu YY. Ceramide-Rubusoside Nanomicelles, a Potential Therapeutic Approach to Target Cancers Carrying p53 Missense Mutations. Mol Cancer Ther 2019; 19:564-574. [PMID: 31645443 DOI: 10.1158/1535-7163.mct-19-0366] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/24/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
Ceramide (Cer) is an active cellular sphingolipid that can induce apoptosis or proliferation-arrest of cancer cells. Nanoparticle-based delivery offers an effective approach for overcoming bioavailability and biopharmaceutics issues attributable to the pronounced hydrophobicity of Cer. Missense mutations of the protein p53, which have been detected in approximately 42% of cancer cases, not only lose the tumor suppression activity of wild-type p53, but also gain oncogenic functions promoting tumor progression and drug resistance. Our previous works showed that cellular Cer can eradicate cancer cells that carry a p53 deletion-mutation by modulating alternative pre-mRNA splicing, restoring wild-type p53 protein expression. Here, we report that new ceramide-rubusoside (Cer-RUB) nanomicelles considerably enhance Cer in vivo bioavailability and restore p53-dependent tumor suppression in cancer cells carrying a p53 missense mutation. Natural RUB encapsulated short-chain C6-Cer so as to form Cer-RUB nanomicelles (∼32 nm in diameter) that substantially enhanced Cer solubility and its levels in tissues and tumors of mice dosed intraperitoneally. Intriguingly, Cer-RUB nanomicelle treatments restored p53-dependent tumor suppression and sensitivity to cisplatin in OVCAR-3 ovarian cancer cells and xenograft tumors carrying p53 R248Q mutation. Moreover, Cer-RUB nanomicelles showed no signs of significant nonspecific toxicity to noncancerous cells or normal tissues, including bone marrow. Furthermore, Cer-RUB nanomicelles restored p53 phosphorylated protein and downstream function to wild-type levels in p53 R172H/+ transgenic mice. Altogether, this study, for the first time, indicates that natural Cer-RUB nanomicelles offer a feasible approach for efficaciously and safely targeting cancers carrying p53 missense mutations.
Collapse
Affiliation(s)
- Sachin K Khiste
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Zhijun Liu
- School of Renewable Natural Resources, Louisiana State University Agricultural Center, Baton Rouge, Louisiana
| | - Kartik R Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Mohammad B Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Salman B Hosain
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Xin Gu
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Sami Nazzal
- Department of Pharmaceutical Sciences, Texas Tech University Health Science Center, Dallas, Texas
| | - Ronald A Hill
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Yong-Yu Liu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana.
| |
Collapse
|
27
|
Ceramide Domains in Health and Disease: A Biophysical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1159:79-108. [DOI: 10.1007/978-3-030-21162-2_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Parveen S, Arjmand F, Tabassum S. Clinical developments of antitumor polymer therapeutics. RSC Adv 2019; 9:24699-24721. [PMID: 35528643 PMCID: PMC9069890 DOI: 10.1039/c9ra04358f] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/18/2019] [Indexed: 01/04/2023] Open
Abstract
Polymer therapeutics encompasses polymer-drug conjugates that are nano-sized, multicomponent constructs already in the clinic as antitumor compounds, either as single agents or in combination with other organic drug scaffolds. Nanoparticle-based polymer-conjugated therapeutics are poised to become a leading delivery strategy for cancer treatments as they exhibit prolonged half-life, higher stability and selectivity, water solubility, longer clearance time, lower immunogenicity and antigenicity and often also specific targeting to tissues or cells. Compared to free drugs, polymer-tethered drugs preferentially accumulate in the tumor sites unlike conventional chemotherapy which does not discriminate between the cancer cells and healthy cells, thereby causing severe side-effects. It is also desirable that the drug reaches its site of action at a particular concentration and the therapeutic dose remains constant over a sufficiently long period of time. This can be achieved by opting for new formulations possessing polymeric systems of drug carriers. However, many challenges still remain unanswered in polymeric drug conjugates which need to be readdressed and therefore, can broaden the scope of this field. This review highlights some of the antitumor polymer therapeutics including polymer-drug conjugates, polymeric micelles, polymeric liposomes and other polymeric nanoparticles that are currently under investigation.
Collapse
Affiliation(s)
- Shazia Parveen
- Chemistry Department, Faculty of Science, Taibah University Yanbu Branch 46423 Yanbu Saudi Arabia +966 504522069
| | - Farukh Arjmand
- Department of Chemistry, Aligarh Muslim University Aligarh-202002 India
| | - Sartaj Tabassum
- Department of Chemistry, Aligarh Muslim University Aligarh-202002 India
| |
Collapse
|
29
|
Nayak R, Meerovich I, Dash AK. Translational Multi-Disciplinary Approach for the Drug and Gene Delivery Systems for Cancer Treatment. AAPS PharmSciTech 2019; 20:160. [PMID: 30968269 DOI: 10.1208/s12249-019-1367-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/11/2019] [Indexed: 01/11/2023] Open
Abstract
Over the last several decades, nanoparticulate delivery systems have emerged as advanced drug and gene delivery tools for cancer therapy. However, their translation into clinical use still poses major challenges. Even though many innovative nanoparticulate approaches have shown very positive results both in vitro and in vivo, few of them have found a place in clinical practice. Possible factors responsible for the existing gap in the translation of nanomedicine to clinical practice may include oversimplification of enhanced permeability and retention effect, lack of correlation between the in vivo animal data vs their translation in human, and challenging multiple biological steps experienced during systemic delivery of nanomedicine. Understanding these challenges and coming up with solutions to overcome them is an important step in effective translation of nanomedicine into clinical practice. This review focuses on advancements in the field of nanomedicine used for anti-cancer therapy, including passive targeting, active targeting, and stimuli-controlled delivery. The review further reveals some of the challenges that are currently faced by pharmaceutical scientists in translation of nanomedicine; these include lack of adequate models for preclinical testing that can predict efficacy in humans, absence of appropriate regulatory guidelines for their approval processes, and difficulty in scale-up of the manufacturing of nanodrug delivery systems. A better understanding of these challenges will help us in filling the gap between the bench and bedside in cancer therapy.
Collapse
|
30
|
Delivering Combination Chemotherapies and Targeting Oncogenic Pathways via Polymeric Drug Delivery Systems. Polymers (Basel) 2019; 11:polym11040630. [PMID: 30959799 PMCID: PMC6523645 DOI: 10.3390/polym11040630] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/22/2019] [Accepted: 03/24/2019] [Indexed: 12/24/2022] Open
Abstract
The side-effects associated with chemotherapy necessitates better delivery of chemotherapeutics to the tumor. Nanoparticles can load higher amounts of drug and improve delivery to tumors, increasing the efficacy of treatment. Polymeric nanoparticles, in particular, have been used extensively for chemotherapeutic delivery. This review describes the efforts made to deliver combination chemotherapies and inhibit oncogenic pathways using polymeric drug delivery systems. Combinations of chemotherapeutics with other drugs or small interfering RNA (siRNA) combinations have been summarized. Special attention is given to the delivery of drug combinations that involve either paclitaxel or doxorubicin, two popular chemotherapeutics in clinic. Attempts to inhibit specific pathways for oncotherapy have also been described. These include inhibition of oncogenic pathways (including those involving HER2, EGFR, MAPK, PI3K/Akt, STAT3, and HIF-1α), augmentation of apoptosis by inhibiting anti-apoptosis proteins (Bcl-2, Bcl-xL, and survivin), and targeting dysregulated pathways such as Wnt/β-catenin and Hedgehog.
Collapse
|
31
|
Xie X, Zhang Y, Li F, Lv T, Li Z, Chen H, Jia L, Gao Y. Challenges and Opportunities from Basic Cancer Biology for Nanomedicine for Targeted Drug Delivery. Curr Cancer Drug Targets 2019; 19:257-276. [DOI: 10.2174/1568009618666180628160211] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/15/2018] [Accepted: 06/22/2018] [Indexed: 12/11/2022]
Abstract
Background:Effective cancer therapy is still a great challenge for modern medical research due to the complex underlying mechanisms of tumorigenesis and tumor metastasis, and the limitations commonly associated with currently used cancer therapeutic options. Nanotechnology has been implemented in cancer therapeutics with immense potential for improving cancer treatment.Objective:Through information about the recent advances regarding cancer hallmarks, we could comprehensively understand the pharmacological effects and explore the mechanisms of the interaction between the nanomaterials, which could provide opportunities to develop mechanism-based nanomedicine to treat human cancers.Methods:We collected related information and data from articles.Results:In this review, we discussed the characteristics of cancer including tumor angiogenesis, abnormalities in tumor blood vessels, uncontrolled cell proliferation markers, multidrug resistance, tumor metastasis, cancer cell metabolism, and tumor immune system that provide opportunities and challenges for nanomedicine to be directed to specific cancer cells and portray the progress that has been accomplished in application of nanotechnology for cancer treatment.Conclusion:The information presented in this review can provide useful references for further studies on developing effective nanomedicine for the treatment of cancer.
Collapse
Affiliation(s)
- Xiaodong Xie
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yingying Zhang
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Fengqiao Li
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Tingting Lv
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Ziying Li
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Haijun Chen
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry; Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350116, China
| |
Collapse
|
32
|
Espinoza SM, Patil HI, San Martin Martinez E, Casañas Pimentel R, Ige PP. Poly-ε-caprolactone (PCL), a promising polymer for pharmaceutical and biomedical applications: Focus on nanomedicine in cancer. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2018.1539990] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Sergio Miguel Espinoza
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada, Legaria 694, Col. Irrigación, 11500 Ciudad de México
| | - Harshal Indrabhan Patil
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Eduardo San Martin Martinez
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada, Legaria 694, Col. Irrigación, 11500 Ciudad de México
| | - Rocio Casañas Pimentel
- CONACYT-Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada, Legaria 694, Col. Irrigación, 11500 Ciudad de México
| | - Pradum Pundlikrao Ige
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| |
Collapse
|
33
|
Yang Z, Song J, Tang W, Fan W, Dai Y, Shen Z, Lin L, Cheng S, Liu Y, Niu G, Rong P, Wang W, Chen X. Stimuli-Responsive Nanotheranostics for Real-Time Monitoring Drug Release by Photoacoustic Imaging. Theranostics 2019; 9:526-536. [PMID: 30809290 PMCID: PMC6376186 DOI: 10.7150/thno.30779] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/04/2018] [Indexed: 01/22/2023] Open
Abstract
Molecular photoacoustic imaging (PA) is a promising technology to understand tumor pathology and guide precision therapeutics. Despite the capability of activatable PA probes to image tumor-specific biomarkers, limitations in their molecular structure hamper them from effective drug delivery and the drug release monitoring. Herein, we developed a perylene diimide (PDI) based theranostic platform that provides noninvasive PA imaging signals to monitor tumor-specific pH-responsive drug release. Methods: we first designed and synthesized an acid-responsive amine-substituted PDI derivative. The pH sensitive properties of the PDI was demonstrated by density functional theory (DFT) calculations, UV-vis experiments and PA studies. The theranostic platform (THPDINs) was fabricated by self-assembly of the acid-responsive PDI, a pH irrelevant IR825 dye, and anti-cancer drug doxorubicin (DOX). The PA properties in various pH environment, drug delivery, cytotoxicity, cell uptake, ratiometric PA imaging and anti-tumor efficacy of the THPDINs were investigated in vitro and in vivo by using U87MG glioma cell line and U87MG tumor model. Results: We found that our designed PDI was sensitive to the tumor specific pH environment, reflected by absorbance shift, PA intensity and aggregation morphology changes in aqueous solution. The as-synthesized pH sensitive PDI acted as a molecular switch in the THPDINs, in which the switch can be triggered in the mild acidic tumor microenvironment to accelerate DOX release. Meanwhile, the DOX release could be monitored by ratiometric PA imaging. Conclusions: We developed a multifunctional PDI based theranostic platform for noninvasive real-time ratiometric PA imaging of tumor acidic pH and monitoring of drug release in living mice simultaneously. This strategy will shed light on the development of smart activatable theranostic nanoplatforms and will significantly advance the application of PA theranostics in biology and medicine.
Collapse
|
34
|
Mao C, Li F, Zhao Y, Debinski W, Ming X. P-glycoprotein-targeted photodynamic therapy boosts cancer nanomedicine by priming tumor microenvironment. Am J Cancer Res 2018; 8:6274-6290. [PMID: 30613297 PMCID: PMC6299702 DOI: 10.7150/thno.29580] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/25/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer nanomedicines only modestly improve the overall survival of patients because their anticancer activity is limited by biological barriers posed by the tumor microenvironment. Currently, all the drugs in FDA-approved cancer nanomedicines are substrates for P-glycoprotein (Pgp), and thus, Pgp-mediated multidrug resistance (MDR) remains a hurdle for cancer nanomedicines. Methods: In this study, Pgp-targeted photodynamic therapy (PDT) was developed to enhance the anticancer efficacy of nanomedicines by depleting MDR cancer cells as well as enhancing tumor penetration of nanomedicines. We first examined the Pgp specificity of our targeted PDT approach, and then tested combination therapy of PDT with Doxil in mixed tumor models of MDR cancer cells and stromal cells, mimicking human heterogeneous tumors. Results: In vitro studies showed that the antibody-photosensitizer conjugates produced Pgp-specific cytotoxicity towards MDR cancer cells upon irradiation with a near-infrared light. The studies with a co-culture model of MDR cancer cells and stromal cells revealed synergistic effects in the combination therapy of PDT with Doxil. Using a mouse model of mixed tumors containing MDR cancer cells and stroma cells, we observed markedly enhanced tumor delivery of Doxil after PDT in vivo. We further examined the effects of the two modalities on individual cell populations and their synergism using an in vivo dual substrate bioluminescence assay. The results indicated that Pgp-targeted PDT specifically depleted MDR cancer cells and further enhanced Doxil's actions on both MDR cancer cells and stromal cells. Conclusion: We conclude that our targeted PDT approach markedly enhances anticancer actions of nanomedicines by depleting MDR cancer cells and increasing their tumor penetration, and thereby, may provide an effective approach to facilitate translation of cancer nanomedicines.
Collapse
|
35
|
Preparation of Magnetic Nanoliposomes of Sesquiterpene-Rich Fraction from Cichorium glandulosum and Its Tissue Distribution in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8549519. [PMID: 30420898 PMCID: PMC6211204 DOI: 10.1155/2018/8549519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/19/2018] [Indexed: 11/30/2022]
Abstract
Objective To prepare and study the distribution of magnetic nanoliposomes of Sesquiterpene-Rich Fraction from Cichorium glandulosum (SRF-MLN) (under magnetic field) in the main organs of mice. Methods The SRF-MLN was prepared by ethanol injection-sonication precipitation method. The quality of its pharmaceutical properties was investigated by the active ingredient lactucin. The drug concentration and distribution of lactucin in different tissues and organs including whole blood, liver, heart, spleen, lung, and kidney were evaluated with Sesquiterpene-Rich Fraction from Cichorium glandulosum (SRF-LP) as a control. Results The prepared SRF-MLN was spherical and monodispersed with an average particle diameter of 65 ± 1 nm, encapsulation efficiency of 91.3% ± 1.5% (RSD% = 1.5), and drug loading rate of 4.7% ± 0.3% (RSD% = 0.7). Dispersion coefficient PDI was 0.157 and Zeta potential was −17.5 mV. The lactucin concentration of SRF-MLN after 10 min of intravenous injection in heart, liver, spleen, lung, and kidney was significantly higher than that of SRF-LP group. The AUC0-12h values of liver tissue were obviously higher than other tissues and MRT was significantly prolonged under the action of the magnetic field (p<0.01). Conclusion The prepared SRF-MLN can change the distribution of drugs in different tissues and organs, prolonging the action time of the drugs in the body, and it has certain specificity under the action of applied magnetic field.
Collapse
|
36
|
Kreitzburg KM, van Waardenburg RCAM, Yoon KJ. Sphingolipid metabolism and drug resistance in ovarian cancer. ACTA ACUST UNITED AC 2018; 1:181-197. [PMID: 31891125 PMCID: PMC6936734 DOI: 10.20517/cdr.2018.06] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite progress in understanding molecular aberrations that contribute to the development and progression of ovarian cancer, virtually all patients succumb to drug resistant disease at relapse. Emerging data implicate bioactive sphingolipids and regulation of sphingolipid metabolism as components of response to chemotherapy or development of resistance. Increases in cytosolic ceramide induce apoptosis in response to therapy with multiple classes of chemotherapeutic agents. Aberrations in sphingolipid metabolism that accelerate the catabolism of ceramide or that prevent the production and accumulation of ceramide contribute to resistance to standard of care platinum- and taxane-based agents. The aim of this review is to highlight current literature and research investigating the influence of the sphingolipids and enzymes that comprise the sphingosine-1-phosphate pathway on the progression of ovarian cancer. The focus of the review is on the utility of sphingolipid-centric therapeutics as a mechanism to circumvent drug resistance in this tumor type.
Collapse
Affiliation(s)
- Kelly M Kreitzburg
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Karina J Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
37
|
Singh SK, Lillard JW, Singh R. Reversal of drug resistance by planetary ball milled (PBM) nanoparticle loaded with resveratrol and docetaxel in prostate cancer. Cancer Lett 2018; 427:49-62. [PMID: 29678549 DOI: 10.1016/j.canlet.2018.04.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/06/2018] [Accepted: 04/15/2018] [Indexed: 01/28/2023]
Abstract
The folate receptor (FR) is a valued target that is highly expressed in various cancers, which will expedite the development of ligand-receptor binding based cancer therapeutics. In the present investigation, through tissue microarray analysis, we report higher levels of folate receptor expression in prostate cancer (PCa) tissue derived from patients, which were minimal in normal tissue. For folate-receptor based targeted therapy of PCa, we generated novel planetary ball milled (PBM) nanoparticles (NPs) encapsulated with resveratrol (RES), and in combination with docetaxel (DTX) and conjugated with folic acid (FA) on the surface. The cytotoxic effect of FA-conjugated DTX-nanoparticles was found effectual that reduced the concentration of free drug (DTX) to 28 times. Flow cytometry analysis showed a significant increase in the number of apoptotic cells by 30.92% and 65.9% in the FA-conjugated RES and in combination with DTX nanoparticle formulation respectively. However, only 8.9% apoptotic cells were found with control (empty NP). The expressions of NF-kB p65, COX-2, pro (BAX, BAK) and anti-apoptotic (BCL-2, BCL-XL) genes were significantly reduced after treatment with FA-RES + DTX-NP. In addition, the FA-conjugated DTX formulation exhibited additional cytotoxic effects with the down-regulation of survivin and an increased expression of Cleaved Caspase-3 in PCa cells. Further, we observed that treating DTX resistant PCa cells with FA-RES + DTX-NP exhibited a reversal of the ABC-transporter markers thereby limiting the multidrug resistance phenotype of the cancer cells. Our results strongly suggested that FA conjugated nanoparticle drugs acted as effective inhibitors of drug efflux that effectually enhances the intracellular concentration of the drug to exhibit their cytotoxic effect.
Collapse
Affiliation(s)
- Santosh Kumar Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - James W Lillard
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| |
Collapse
|
38
|
Yang H, Guo J, Tong R, Yang C, Chen JK. pH-Sensitive Micelles Based on Star Copolymer Ad-(PCL-b-PDEAEMA-b-PPEGMA)₄ for Controlled Drug Delivery. Polymers (Basel) 2018; 10:E443. [PMID: 30966478 PMCID: PMC6415201 DOI: 10.3390/polym10040443] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/07/2018] [Accepted: 04/11/2018] [Indexed: 02/01/2023] Open
Abstract
Enhancing drug loading efficacy and stability of polymeric micelles remains a grand challenge. Here we develop adamantane-based star copolymers adamantane-[poly(ε-caprolactone)-b-poly(2-(diethylamino)ethyl methacrylate)-b-poly(poly(ethylene glycol) methyl ether methacrylate)]₄ (Ad-(PCL-b-PDEAEMA-b-PPEGMA)₄) and their self-assembled micelles for controlled drug delivery. Results show that the polymers have excellent stability in solution with low critical micelle concentration (CMC) (0.0025⁻0.0034 mg/mL) and the apparent base dissociation constant (pKb) of the polymers is from 5.31 to 6.05. Dynamic light scattering analysis exhibits the great environmental response capability of the pH-sensitive micelles according to particle sizes and zeta potentials. With the synergy effect of the adamantane and hydrophobic block, the micelles display the high Doxorubicin (DOX) loading efficacy (up to 22.4%). The DOX release study shows that the micelles are capable of controlled release for drug. This work indicates the Ad-(PCL-b-PDEAEMA-b-PPEGMA)₄ micelles may provide new guidelines for drug control and release system in overcoming cancer treatment.
Collapse
Affiliation(s)
- Huiyan Yang
- School of Chemical Engineering & Light Industry, Guangdong University of Technology, Guangzhou 510006, China.
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, No. 43, Sec. 4, Keelung Road, Taipei 106, Taiwan.
| | - Jianwei Guo
- School of Chemical Engineering & Light Industry, Guangdong University of Technology, Guangzhou 510006, China.
| | - Rui Tong
- Guangzhou Tinci Materials Technology Co., Ltd., Guangzhou 510760, China.
| | - Chufen Yang
- School of Chemical Engineering & Light Industry, Guangdong University of Technology, Guangzhou 510006, China.
| | - Jem-Kun Chen
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, No. 43, Sec. 4, Keelung Road, Taipei 106, Taiwan.
| |
Collapse
|
39
|
Ahmad G, Gattacecca F, El Sadda R, Botchkina G, Ojima I, Egan J, Amiji M. Biodistribution and Pharmacokinetic Evaluations of a Novel Taxoid DHA-SBT-1214 in an Oil-in-Water Nanoemulsion Formulation in Naïve and Tumor-Bearing Mice. Pharm Res 2018; 35:91. [PMID: 29520477 PMCID: PMC6151135 DOI: 10.1007/s11095-018-2349-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/17/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE The main purpose of this study was to formulate an oil-in-water nanoemulsion of a next generation taxoid DHA-SBT-1214 and evaluate its biodistribution and pharmacokinetics. METHODS DHA-SBT-1214 was encapsulated in a fish oil containing nanoemulsion using a high pressure homogenization method. Following morphological characterization of the nanoemulsions, qualitative and quantitative biodistribution was evaluated in naïve and cancer stem cell-enriched PPT-2 human prostate tumor bearing mice. RESULTS DHA-SBT-1214 was successfully encapsulated up to 20 mg/ml in the nanoemulsion formulation and had an average oil droplet size of 200 nm. Using a DiR near infra-red dye encapsulated nanoemulsion, we have shown the delivery of nanoemulsion to mouse tumor region. By quantitative analysis, DHA-SBT-1214 encapsulated nanoemulsion demonstrated improved pharmacokinetic properties in plasma and different tissues as compared to its solution form. Furthermore, the nanoemulsions were stable and had slower in vitro drug release compared to its solution form. CONCLUSIONS The results from this study demonstrated effective encapsulation of the drug in a nanoemulsion and this nanoemulsion showed sustained plasma levels and enhanced tumor delivery relative to the solution form.
Collapse
Affiliation(s)
- Gulzar Ahmad
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Massachusetts, 02115-5000, USA
| | - Florence Gattacecca
- Institut de Recherche en Cancérologie de Montpellier IRCM, INSERM U1194, ICM, Université de Montpellier, Montpellier, France
| | - Rana El Sadda
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, 11794-3400, USA
| | - Galina Botchkina
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, 11794-3400, USA
- Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, New York, 11794-8691, USA
| | - Iwao Ojima
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York, 11794-3400, USA
- Department of Chemistry, Stony Brook University, Stony Brook, New York, 11794-3400, USA
| | - James Egan
- Targagenix, Inc., 25 Health Sciences Drive, Stony Brook, New York, 11790-3382, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Massachusetts, 02115-5000, USA.
| |
Collapse
|
40
|
Lin J, Zhao C, Liu C, Fu S, Han L, Lu X, Yang C. Redox-responsive F127-folate/F127-disulfide bond-d-α-tocopheryl polyethylene glycol 1000 succinate/P123 mixed micelles loaded with paclitaxel for the reversal of multidrug resistance in tumors. Int J Nanomedicine 2018; 13:805-830. [PMID: 29445276 PMCID: PMC5808690 DOI: 10.2147/ijn.s152395] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Introduction The development of nanodrug carriers utilizing tumor microenvironment has become a hotspot in reversing multidrug resistance (MDR). Materials and methods This study synthesized a redox-sensitive copolymer, Pluronic F127-disulfide bond-d-α-tocopheryl polyethylene glycol 1000 succinate (FSST), through the connection of the reduction-sensitive disulfide bond between F127 and d-α-tocopheryl polyethylene glycol 1000 succinate. This polymer could induce the elevation of reactive oxygen species (ROS) levels, ultimately resulting in cytotoxicity. Moreover, the redox-responsive mixed micelles, F127-folate (FA)/FSST/P123 (FFSSTP), based on FSST, Pluronic F127-FA, and Pluronic P123, were prepared to load paclitaxel (PTX). Results The in vitro release study demonstrated that FFSSTP/PTX accelerated the PTX release through the breakage of disulfide bond in reductive environment. In cellular experiment, FFSSTP/PTX induced significant apoptosis in PTX-resistant MCF-7/PTX cells through inhibiting adenosine triphosphate (ATP)-binding cassette proteins from pumping out PTX by interfering with the mitochondrial function and ATP synthesis. Furthermore, FFSSTP/PTX induced apoptosis through elevating the intracellular levels of ROS. Conclusion FFSSTP could become a potential carrier for the treatment of MDR tumors.
Collapse
Affiliation(s)
- Jing Lin
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Chaoyue Zhao
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Cuijuan Liu
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Shiyao Fu
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Luying Han
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Xinping Lu
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Chunrong Yang
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| |
Collapse
|
41
|
Rodríguez-Alcalá LM, Castro-Gómez MP, Pimentel LL, Fontecha J. Milk fat components with potential anticancer activity-a review. Biosci Rep 2017; 37:BSR20170705. [PMID: 29026007 PMCID: PMC6372256 DOI: 10.1042/bsr20170705] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 08/04/2017] [Accepted: 10/02/2017] [Indexed: 02/07/2023] Open
Abstract
During many years, the milk fat has been unfairly undervalued due to its association with higher levels of cardiovascular diseases, dyslipidaemia or obesity, among others. However, currently, this relationship is being re-evaluated because some of the dairy lipid components have been attributed potential health benefits. Due to this, and based on the increasing incidence of cancer in our society, this review work aims to discuss the state of the art concerning scientific evidence of milk lipid components and reported anticancer properties. Results from the in vitro and in vivo experiments suggest that specific fatty acids (FA) (as butyric acid and conjugated linoleic acid (CLA), among others), phospholipids and sphingolipids from milk globule membrane are potential anticarcinogenic agents. However, their mechanism of action remains still unclear due to limited and inconsistent findings in human studies.
Collapse
Affiliation(s)
- Luis M Rodríguez-Alcalá
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Arquiteto Lobão Vital, Apartado 2511, Porto 4202-401, Portugal
- Research Center for Natural Resources and Sustainability (CIRENYS), Bernardo O'Higgins University, Fábrica N° 1990, Segundo Piso, Santiago de Chile, Chile
| | - M Pilar Castro-Gómez
- Institute of Food Science Research, (CIAL, CSIC-UAM), Department of Bioactivity and Food Analysis, Food Lipid Biomarkers and Health Group, Campus of Autónoma University of Madrid, C/Nicolás Cabrera, Madrid 9. 28049, Spain
| | - Lígia L Pimentel
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Arquiteto Lobão Vital, Apartado 2511, Porto 4202-401, Portugal
| | - Javier Fontecha
- Institute of Food Science Research, (CIAL, CSIC-UAM), Department of Bioactivity and Food Analysis, Food Lipid Biomarkers and Health Group, Campus of Autónoma University of Madrid, C/Nicolás Cabrera, Madrid 9. 28049, Spain
| |
Collapse
|
42
|
Chen C, Jia F, Hou Z, Ruan S, Lu Q. Delivery of paeonol by nanoparticles enhances its in vitro and in vivo antitumor effects. Int J Nanomedicine 2017; 12:6605-6616. [PMID: 28924345 PMCID: PMC5595364 DOI: 10.2147/ijn.s143938] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Paeonol (Pae; 2′-hydroxy-4′-methoxyacetophenone) has attracted intense attention as a potential therapeutic agent against various cancers. However, the use of Pae is limited owing to its hydrophobicity. Recently, biodegradable polymeric nanoparticles with amphiphilic copolymers have been used as drug carriers; these have better bioavailability and are promising tumor-targeted drug delivery systems. In the current study, we prepared Pae-loaded nanoparticles (Pae-NPs) with amphiphilic block copolymers using nanoprecipitation. The physiochemical characteristics and antitumor effects of nanoparticles were evaluated in different cancer cells. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays showed substantial inhibition of cell growth by Pae-NPs. Moreover, lower doses of Pae-NPs inhibited cell growth more efficiently than the equivalent doses of free Pae. Inhibition was characterized by significant elevation of intracellular reactive oxygen species and subsequent inhibition of Akt and regulation of apoptotic proteins, which could be partly reversed by pretreatment with the antioxidant N-acetylcysteine. In vivo results also demonstrated that Pae-NPs could exert much stronger antitumor effects than free Pae. Therefore, Pae-NPs represent a promising delivery system to overcome the low solubility of Pae and enable its use in treating cancer.
Collapse
Affiliation(s)
- Cong Chen
- Department of Gynecology of Traditional Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing
| | - Feng Jia
- Department of Neurosurgery, Yancheng City No 1 People's Hospital, The Fourth Affiliated Hospital of Nantong Medical College, Yancheng
| | - Zhibo Hou
- First Department of Respiratory Medicine, Nanjing Chest Hospital, Medical School of Southeast University, Nanjing
| | - Shu Ruan
- Department of Endocrinology, Yancheng Third Hospital, The Affiliated Hospital of Southeast University Medical College, Yancheng, Jiangsu, China
| | - Qibin Lu
- Department of Gynecology of Traditional Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing
| |
Collapse
|
43
|
Øverbye A, Holsæter AM, Markus F, Škalko-Basnet N, Iversen TG, Torgersen ML, Sønstevold T, Engebraaten O, Flatmark K, Mælandsmo GM, Skotland T, Sandvig K. Ceramide-containing liposomes with doxorubicin: time and cell-dependent effect of C6 and C12 ceramide. Oncotarget 2017; 8:76921-76934. [PMID: 29100358 PMCID: PMC5652752 DOI: 10.18632/oncotarget.20217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/17/2017] [Indexed: 12/14/2022] Open
Abstract
Doxorubicin, a widely used chemotherapeutic drug, has several potential high-risk side effects including cardiomyopathy. Furthermore, cellular resistance to this drug develops with time. By using liposomes as carrier vesicles both the side effects and drug resistance might be avoided. In this study we have investigated the cytotoxic effect of doxorubicin encapsulated in liposomes with and without ceramides containing 6 or 12 carbon atoms in the N-amidated fatty acyl chains. The short-chain ceramide species were included in the liposomal compositions due to their pro-apoptotic properties, which might cause a synergistic anticancer effect. We demonstrate that the ceramide species enhance the liposomal doxorubicin toxicity in a cell-specific manner. The C6-ceramide effect is most pronounced in cervical cancer cells (HeLa) and colon cancer cells (HCT116), whereas the C12-ceramide effect is strongest in breast cancer cells (MDA-MB-231). Moreover, the study reveals the importance of investigating cell toxicity at several time points and in different cell-lines, to assess drug-and formulation-induced cytotoxic effects in vitro. Furthermore, our data show that the cytotoxicity obtained with the nanocarriers in vitro, does not necessarily reflect their ability to inhibit tumor growth in vivo. We speculate that the larger effect of Caelyx® than our liposomes in vivo is due to a greater in vivo stability of Caelyx®.
Collapse
Affiliation(s)
- Anders Øverbye
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ann Mari Holsæter
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Fusser Markus
- Department of Tumour Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Tore-Geir Iversen
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Tonje Sønstevold
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Olav Engebraaten
- Department of Tumour Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjersti Flatmark
- Department of Tumour Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gunhild Mari Mælandsmo
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, Tromsø, Norway.,Department of Tumour Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Tore Skotland
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Centre for Cancer Biomedicine, Faculty Division Norwegian Radium Hospital, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
44
|
Nanoemulsion formulation of a novel taxoid DHA-SBT-1214 inhibits prostate cancer stem cell-induced tumor growth. Cancer Lett 2017; 406:71-80. [PMID: 28803993 DOI: 10.1016/j.canlet.2017.08.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/20/2017] [Accepted: 08/03/2017] [Indexed: 02/01/2023]
Abstract
The main aim of this study was to evaluate the therapeutic efficacy of an oil-in-water nanoemulsion formulation encapsulating DHA-SBT-1214, a novel omega-3 fatty acid conjugated taxoid prodrug, against prostate cancer stem cells. Nanoemulsions of DHA-SBT-1214 (NE-DHA-SBT-1214) were prepared and characterized. In vitro delivery efficiency and cytotoxicity of NE-DHA-SBT-1214 was compared with solution formulation in PPT2 cells. In vivo studies included analysis of comparative efficacy of NE-DHA-SBT-1214 with Abraxane® and placebo nanoemulsions as well as post-treatment alternations in clonogenic and sphere-forming capabilities of the tumor cells. Qualitative intracellular uptake studies of dye encapsulated NEs by confocal imaging showed uptake by both monolayer and spheroid cultured PPT2 cells. Treatment of PPT2 cells with NE DHA-SBT-1214 (1nM-1μM for monolayer culture of cells grown on collagen-coated dishes for 48 h) induced complete cell death, showing higher efficacy as compared to the drug solution. This nanoemulsion (10nM-10μM) also showed toxicity in 3D culture of floating spheroids. Weekly intravenous administration of the NE-DHA-SBT-1214 to NOD/SCID mice bearing subcutaneous PPT2 tumor xenografts led to dramatic suppression of tumor growth compared to Abraxane® and placebo nanoemulsion formulation. Viable cells that survived from this in vivo treatment regimen were no longer able to induce floating spheroids and holoclones, whereas control and Abraxane® treated tumor cells induced a large number of both. The results show that NE-DHA-SBT-1214 possesses significant activity against prostate CD133high/CD44+/high tumor-initiating cells both in vitro and in vivo.
Collapse
|
45
|
Singh MS, Tammam SN, Shetab Boushehri MA, Lamprecht A. MDR in cancer: Addressing the underlying cellular alterations with the use of nanocarriers. Pharmacol Res 2017; 126:2-30. [PMID: 28760489 DOI: 10.1016/j.phrs.2017.07.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/29/2017] [Accepted: 07/26/2017] [Indexed: 01/02/2023]
Abstract
Multidrug resistance (MDR) is associated with a wide range of pathological changes at different cellular and intracellular levels. Nanoparticles (NPs) have been extensively exploited as the carriers of MDR reversing payloads to resistant tumor cells. However, when properly formulated in terms of chemical composition and physicochemical properties, NPs can serve as beyond delivery systems and help overcome MDR even without carrying a load of chemosensitizers or MDR reversing molecular cargos. Whether serving as drug carriers or beyond, a wise design of the nanoparticulate systems to overcome the cellular and intracellular alterations underlying the resistance is imperative. Within the current review, we will initially discuss the cellular changes occurring in resistant cells and how such changes lead to chemotherapy failure and cancer cell survival. We will then focus on different mechanisms through which nanosystems with appropriate chemical composition and physicochemical properties can serve as MDR reversing units at different cellular and intracellular levels according to the changes that underlie the resistance. Finally, we will conclude by discussing logical grounds for a wise and rational design of MDR reversing nanoparticulate systems to improve the cancer therapeutic approaches.
Collapse
Affiliation(s)
- Manu S Singh
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany
| | - Salma N Tammam
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Department of Pharmaceutical Technology, German University of Cairo, Egypt
| | | | - Alf Lamprecht
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Laboratory of Pharmaceutical Engineering (EA4267), University of Franche-Comté, Besançon, France.
| |
Collapse
|
46
|
Dany M. Sphingosine metabolism as a therapeutic target in cutaneous melanoma. Transl Res 2017; 185:1-12. [PMID: 28528915 DOI: 10.1016/j.trsl.2017.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/26/2017] [Accepted: 04/25/2017] [Indexed: 12/19/2022]
Abstract
Melanoma is by far the most aggressive type of skin cancer with a poor prognosis in its advanced stages. Understanding the mechanisms involved in melanoma pathogenesis, response, and resistance to treatment has gained a lot of attention worldwide. Recently, the role of sphingolipid metabolism has been studied in cutaneous melanoma. Sphingolipids are bioactive lipid effector molecules involved in the regulation of various cellular signaling pathways such as inflammation, cancer cell proliferation, death, senescence, and metastasis. Recent studies suggest that sphingolipid metabolism impacts melanoma pathogenesis and is a potential therapeutic target. This review focuses on defining the role of sphingolipid metabolism in melanoma carcinogenesis, discussing sphingolipid-based therapeutic approaches, and highlighting the areas that require more extensive research.
Collapse
Affiliation(s)
- Mohammed Dany
- College of Medicine, Medical University of South Carolina, Charleston, SC.
| |
Collapse
|
47
|
Yuan Y, Xu L, Dai S, Wang M, Wang H. A facile supramolecular approach to fabricate multifunctional upconversion nanoparticles as a versatile platform for drug loading, in vivo delivery and tumor imaging. J Mater Chem B 2017; 5:2425-2435. [PMID: 32264550 DOI: 10.1039/c6tb03381d] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multifunctional upconversion nanoparticles (UCNPs) that can be implemented in theranostic applications are particularly attractive scaffolds for precise drug delivery. However, most of the current methods for drug formulation are technically complicated, thereby impeding their use in the clinic. Here, we report on the preparation of a lipophilic cytotoxic prodrug-integrated and polyethylene glycol (PEG)-cloaked UCNPs scaffold through a facile one-pot supramolecular approach. By choosing 7-ethyl-10-hydroxycamptothecin (SN38)-derived prodrug 1 as a model chemotherapeutic, we show that this lipophilic prodrug can be feasibly self-assembled onto the surface of UCNPs, which are cooperatively solubilized by PEGylated phospholipids. The resulting SN38 prodrug 1-encapsulated UCNPs (designated 1@pUCNPs) produce a stable colloidal system in aqueous solution, making it suitable for intravenous injection. The SN38 drug loading capacity in pUCNPs is as high as ∼12.3 wt%, and a sustained drug release profile is observed, indicating that the drug payloads can be transported to targeted tumor sites via the enhanced permeability and retention (EPR) effect. Upconversion luminescence (UCL) imaging, including in vivo and ex vivo imaging, suggests that the drug-loaded pUCNPs remain stable in tumors over a long time and preferentially accumulate in tumors presumably via the EPR effect. Furthermore, the 1@pUCNPs show superior therapeutic outcomes compared with the clinically approved SN38 prodrug CPT-11 in the Bcap-37 mouse model of breast cancer. Collectively, our results demonstrate that pUCNPs facilely constructed in a one-pot self-assembly manner may be used as a versatile platform, enabling synchronous in vivo delivery of poorly water-soluble drugs and tumor imaging.
Collapse
Affiliation(s)
- Yingying Yuan
- The First Affiliated Hospital, School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University, Hangzhou 310003, P. R. China.
| | | | | | | | | |
Collapse
|
48
|
Cell membrane modulation as adjuvant in cancer therapy. Cancer Treat Rev 2016; 52:48-57. [PMID: 27889637 DOI: 10.1016/j.ctrv.2016.10.008] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/24/2016] [Accepted: 10/27/2016] [Indexed: 12/25/2022]
Abstract
Cancer is a complex disease involving numerous biological processes, which can exist in parallel, can be complementary, or are engaged when needed and as such can replace each other. This redundancy in possibilities cancer cells have, are fundamental to failure of therapy. However, intrinsic features of tumor cells and tumors as a whole provide also opportunities for therapy. Here we discuss the unique and specific makeup and arrangement of cell membranes of tumor cells and how these may help treatment. Interestingly, knowledge on cell membranes and associated structures is present already for decades, while application of membrane modification and manipulation as part of cancer therapy is lagging. Recent developments of scientific tools concerning lipids and lipid metabolism, opened new and previously unknown aspects of tumor cells and indicate possible differences in lipid composition and membrane function of tumor cells compared to healthy cells. This field, coined Lipidomics, demonstrates the importance of lipid components in cell membrane in several illnesses. Important alterations in cancer, and specially in resistant cancer cells compared to normal cells, opened the door to new therapeutic strategies. Moreover, the ability to modulate membrane components and/or properties has become a reality. Here, developments in cancer-related Lipidomics and strategies to interfere specifically with cancer cell membranes and how these affect cancer treatment are discussed. We hypothesize that combination of lipid or membrane targeted strategies with available care to improve chemotherapy, radiotherapy and immunotherapy will bring the much needed change in treatment in the years to come.
Collapse
|
49
|
Abstract
Development of novel drug-delivery systems aims to specifically deliver anticancer drugs to tumor tissues and improve the efficiency of chemotherapy, while minimizing side effects of drugs on healthy tissues and organs. However, drug-delivery systems are confronted by membrane barriers and multiple drug resistance in cancer cells. In recent years, the obtained results indicate an important role of lipids, proteins and carbohydrates in apoptosis, drug transport and the process of cellular uptake of nanoparticles via endocytosis. This article discusses the hypothesis of the relationship between cell membrane structure and nanoparticles in cancer cells.
Collapse
|
50
|
Patel A, Sant S. Hypoxic tumor microenvironment: Opportunities to develop targeted therapies. Biotechnol Adv 2016; 34:803-812. [PMID: 27143654 PMCID: PMC4947437 DOI: 10.1016/j.biotechadv.2016.04.005] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/13/2016] [Accepted: 04/28/2016] [Indexed: 01/18/2023]
Abstract
In recent years, there has been great progress in the understanding of tumor biology and its surrounding microenvironment. Solid tumors create regions with low oxygen levels, generally termed as hypoxic regions. These hypoxic areas offer a tremendous opportunity to develop targeted therapies. Hypoxia is not a random by-product of the cellular milieu due to uncontrolled tumor growth; rather it is a constantly evolving participant in overall tumor growth and fate. This article reviews current trends and recent advances in drug therapies and delivery systems targeting hypoxia in the tumor microenvironment. In the first part, we give an account of important physicochemical changes and signaling pathways activated in the hypoxic microenvironment. This is then followed by various treatment strategies including hypoxia-sensitive signaling pathways and approaches to develop hypoxia-targeted drug delivery systems.
Collapse
Affiliation(s)
- Akhil Patel
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, United States
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|