1
|
Datta D, Sulthana S, Strauss J, Puri A, Priyanka Bandi S, Singh S. Reconnoitring signaling pathways and exploiting innovative approaches tailoring multifaceted therapies for skin cancer. Int J Pharm 2024; 665:124719. [PMID: 39293575 DOI: 10.1016/j.ijpharm.2024.124719] [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: 06/08/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Nowadays, skin cancer is widespread just like a varied malignant cancer which can cause serious health issues. Skin cancer, which encompasses malignant melanoma, basal cell carcinoma, and squamous cell carcinoma, is a prevalent form of cancer among humans. Due to its broad prevalence, financial burden, mortality rates, and cosmetic effects, it is a major public health issue. Skin cancer treatment involves surgery, chemotherapy, and radiation. Recently, personalized treatment in the fields of targeted therapies and precision medicine has been shown to diagnose early detection of every individual tumor by knowing their genetic and molecular characteristics. To target the molecular pathways responsible for tumor growth and reduce the damage to healthy tissue, new targeted therapies have emerged for melanoma, basal cell carcinoma, and squamous cell carcinoma. B-raf serine/threonine kinase (BRAF) and mitogen-activated protein kinase (MEK) inhibitors, immune checkpoint inhibitors, and precision medications have strong response rates to improve patient survival. Targeted therapeutics like nanocarriers have shown promising results by reducing skin irritation and protecting encapsulated therapeutics. These formulations have been shown to improve the transdermal permeability of anticancer drugs. The consideration of employing physical techniques to enhance the permeation of nanocarriers warrants attention to augment the dermal permeation of anticancer agents and facilitate targeted drug delivery within neoplastic cells. Targeted therapies face obstacles like resistance mechanisms and treatment strategy monitoring. Taken together, this review delves into the basic mechanisms of skin cancer, current treatment methods, drug resistance processes, and nano-based targeted techniques for cancer treatment. It will also delineate the challenges and perspectives in pre-clinical and clinical contexts.
Collapse
Affiliation(s)
- Deepanjan Datta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India.
| | - Safiya Sulthana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Jordan Strauss
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614
| | - Sony Priyanka Bandi
- Loka Laboratories Private Limited, Technology Business Incubator, BITS Pilani Hyderabad Campus, Jawahar Nagar, Medchal 500078, Telangana, India.
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
2
|
Honfroy A, Bertouille J, Turea AM, Cauwenbergh T, Bridoux J, Lensen N, Mangialetto J, Van den Brande N, White JF, Gardiner J, Brigaud T, Ballet S, Hernot S, Chaume G, Martin C. Fluorinated Peptide Hydrogels Result in Longer In Vivo Residence Time after Subcutaneous Administration. Biomacromolecules 2024; 25:6666-6680. [PMID: 39230056 DOI: 10.1021/acs.biomac.4c00872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Peptide-based hydrogels are of interest to biomedical applications. Herein, we have explored the introduction of fluorinated amino acids in hydrogelator H-FQFQFK-NH2 (P1) to design a series of fluorinated peptide hydrogels and evaluate the in vitro and in vivo properties of the most promising analogues. The impact of fluorinated groups on peptide gelation, secondary structure, and self-assembly processes was assessed. We show that fluorine can significantly improve hydrogel stiffness, compared to the nonfluorinated reference P1. For P15 (H-FQFQF(o-CF3)K-NH2), P18 (H-FQFQF(F5)K-NH2), and P19 (H-FQFQM(CF3)K-NH2), microscopy studies scrutinized fiber morphologies and alignment in the network. In vitro release studies of hydrogels loaded with an opioid cargo suggested improved hydrogel stability for P15 and P18. This improved stability was further validated in vivo, notably for P15, giving the most significant increased gel residence time, with more than 20% of hydrogel still present 9 days post-injection, as monitored by nuclear SPECT-CT imaging.
Collapse
Affiliation(s)
- Aurélie Honfroy
- Research Group of Organic Chemistry (ORGC), Vrije Universiteit Brussel, Pleinlaan 2, Brussels B-1050, Belgium
- VUB, Molecular Imaging and Therapy Research Group (MITH), Laarbeeklaan 103, Jette 1090, Belgium
- CY Cergy Paris Université, CNRS, BioCIS UMR 8076, Cergy-Pontoise 95000, France
- Université Paris-Saclay, CNRS, BioCIS UMR 8076, Orsay 91400, France
| | - Jolien Bertouille
- Research Group of Organic Chemistry (ORGC), Vrije Universiteit Brussel, Pleinlaan 2, Brussels B-1050, Belgium
| | - Ana-Maria Turea
- Research Group of Organic Chemistry (ORGC), Vrije Universiteit Brussel, Pleinlaan 2, Brussels B-1050, Belgium
| | - Thibault Cauwenbergh
- Research Group of Organic Chemistry (ORGC), Vrije Universiteit Brussel, Pleinlaan 2, Brussels B-1050, Belgium
| | - Jessica Bridoux
- VUB, Molecular Imaging and Therapy Research Group (MITH), Laarbeeklaan 103, Jette 1090, Belgium
| | - Nathalie Lensen
- CY Cergy Paris Université, CNRS, BioCIS UMR 8076, Cergy-Pontoise 95000, France
- Université Paris-Saclay, CNRS, BioCIS UMR 8076, Orsay 91400, France
| | - Jessica Mangialetto
- Research Group Sustainable Materials Engineering (SUME), Lab of Physical Chemistry and Polymer Science (FYSC), Vrije Universiteit Brussel, Pleinlaan 2, Brussels B-1050, Belgium
| | - Niko Van den Brande
- Research Group Sustainable Materials Engineering (SUME), Lab of Physical Chemistry and Polymer Science (FYSC), Vrije Universiteit Brussel, Pleinlaan 2, Brussels B-1050, Belgium
| | - Jacinta F White
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3169, Australia
| | - James Gardiner
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3169, Australia
| | - Thierry Brigaud
- CY Cergy Paris Université, CNRS, BioCIS UMR 8076, Cergy-Pontoise 95000, France
- Université Paris-Saclay, CNRS, BioCIS UMR 8076, Orsay 91400, France
| | - Steven Ballet
- Research Group of Organic Chemistry (ORGC), Vrije Universiteit Brussel, Pleinlaan 2, Brussels B-1050, Belgium
| | - Sophie Hernot
- VUB, Molecular Imaging and Therapy Research Group (MITH), Laarbeeklaan 103, Jette 1090, Belgium
| | - Grégory Chaume
- CY Cergy Paris Université, CNRS, BioCIS UMR 8076, Cergy-Pontoise 95000, France
- Université Paris-Saclay, CNRS, BioCIS UMR 8076, Orsay 91400, France
| | - Charlotte Martin
- Research Group of Organic Chemistry (ORGC), Vrije Universiteit Brussel, Pleinlaan 2, Brussels B-1050, Belgium
| |
Collapse
|
3
|
Rothe R, Xu Y, Wodtke J, Brandt F, Meister S, Laube M, Lollini PL, Zhang Y, Pietzsch J, Hauser S. Programmable Release of Chemotherapeutics from Ferrocene-Based Injectable Hydrogels Slows Melanoma Growth. Adv Healthc Mater 2024; 13:e2400265. [PMID: 39007274 DOI: 10.1002/adhm.202400265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Hydrogel-based injectable drug delivery systems provide temporally and spatially controlled drug release with reduced adverse effects on healthy tissues. Therefore, they represent a promising therapeutic option for unresectable solid tumor entities. In this study, a peptide-starPEG/hyaluronic acid-based physical hydrogel is modified with ferrocene to provide a programmable drug release orchestrated by matrix-drug interaction and local reactive oxygen species (ROS). The injectable ROS-responsive hydrogel (hiROSponse) exhibits adequate biocompatibility and biodegradability, which are important for clinical applications. HiROSponse is loaded with the two cytostatic drugs (hiROSponsedox/ptx) doxorubicin (dox) and paclitaxel (ptx). Dox is a hydrophilic compound and its release is mainly controlled by Fickian diffusion, while the hydrophobic interactions between ptx and ferrocene can control its release and thus be regulated by the oxidation of ferrocene to the more hydrophilic state of ferrocenium. In a syngeneic malignant melanoma-bearing mouse model, hiROSponsedox/ptx slows tumor growth without causing adverse side effects and doubles the relative survival probability. Programmable release is further demonstrated in a tumor model with a low physiological ROS level, where dox release, low dose local irradiation, and the resulting ROS-triggered ptx release lead to tumor growth inhibition and increased survival.
Collapse
Affiliation(s)
- Rebecca Rothe
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Yong Xu
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Johanna Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Florian Brandt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Sebastian Meister
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Pier-Luigi Lollini
- Alma Mater Studiorum, University of Bologna, Department of Medical and Surgical Sciences, Viale Filopanti 22, Bologna, 40126, Italy
| | - Yixin Zhang
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| |
Collapse
|
4
|
Cao S, Wei Y, Yue Y, Wang D, Yang J, Xiong A, Zeng H. Mapping the evolution and research landscape of ferroptosis-targeted nanomedicine: insights from a scientometric analysis. Front Pharmacol 2024; 15:1477938. [PMID: 39386034 PMCID: PMC11461269 DOI: 10.3389/fphar.2024.1477938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Objective Notable progress has been made in "ferroptosis-based nano drug delivery systems (NDDSs)" over the past 11 years. Despite the ongoing absence of a comprehensive scientometric overview and up-to-date scientific mapping research, especially regarding the evolution, critical research pathways, current research landscape, central investigative themes, and future directions. Methods Data ranging from 1 January 2012, to 30 November 2023, were obtained from the Web of Science database. A variety of advanced analytical tools were employed for detailed scientometric and visual analyses. Results The results show that China significantly led the field, contributing 82.09% of the total publications, thereby largely shaping the research domain. Chen Yu emerged as the most productive author in this field. Notably, the journal ACS Nano had the greatest number of relevant publications. The study identified liver neoplasms, pancreatic neoplasms, gliomas, neoplasm metastases, and melanomas as the top five crucial disorders in this research area. Conclusion This research provides a comprehensive scientometric assessment, enhancing our understanding of NDDSs focused on ferroptosis. Consequently, it enables rapid access to essential information and facilitates the extraction of novel ideas in the field of ferroptotic nanomedicine for both experienced and emerging researchers.
Collapse
Affiliation(s)
- Siyang Cao
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yihao Wei
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, Hong Kong, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, China
| | - Yaohang Yue
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Deli Wang
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Ao Xiong
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hui Zeng
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Orthopedics, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
5
|
Zhou Q, Xie D, Wang K, Wang F, Wang Q, Huang Y, Yu M, Huang J, Zhao Y. Evodiamine encapsulated by hyaluronic acid modified zeolitic imidazolate framework-8 for tumor targeted therapy. Drug Deliv Transl Res 2024:10.1007/s13346-024-01652-4. [PMID: 38941037 DOI: 10.1007/s13346-024-01652-4] [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] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
Evodiamine (EVO), a natural bioactive compound extracted from Evodia rutaecarpa, shows therapeutic ability against malignant melanoma. However, the poor solubility and bioavailability of EVO limit its clinical application. Metal-organic frameworks (MOFs) have shown excellent physical and chemical properties and are widely used as drug delivery systems. Among them, zeolitic imidazolate framework-8 (ZIF-8) is a research popular material because of its unique properties, such as hydrothermal stability, non-toxicity, biocompatibility, and pH sensitivity. In this study, in order to load EVO, a drug carrier that hyaluronic acid (HA) modified zeolitic imidazolate framework-8 (ZIF-8) is synthesized. This drug carrier has shown drug loading with 6.2 ± 0.6%, and the nano drugs (EVO@ZIF-8/HA) have good dispersibility. Owing to the decoration HA of EVO@ZIF-8, the potential of the nano drugs is reversed from the positive charge to the negative charge, which is beneficial to blood circulation in vivo. Furthermore, because the CD44-expressing in tumor cells is excessed, the endocytosis and accumulation of nano drugs in tumor cells are beneficial to improvement. Compared with free EVO, EVO@ZIF-8/HA has shown a significantly improved anti-tumor efficacy in vitro and in vivo. In summary, the drug carrier effectively addresses the challenges that are caused by the strong hydrophobicity and low bioavailability of EVO, thereby targeted tumor therapy of EVO can be achieved.
Collapse
Affiliation(s)
- Qiang Zhou
- Department of Pharmacy, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
| | - Dandan Xie
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Kui Wang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Fengling Wang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Qiaoling Wang
- Department of Pharmacy, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
| | - Yue Huang
- Department of Pharmacy, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
| | - Mengjun Yu
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Jingbin Huang
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China.
| | - Yu Zhao
- Department of Pharmacy, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China.
| |
Collapse
|
6
|
Gupta N, Gupta G, Razdan K, Albekairi NA, Alshammari A, Singh D. Development of nanoemulgel of 5-Fluorouracil for skin melanoma using glycyrrhizin as a penetration enhancer. Saudi Pharm J 2024; 32:101999. [PMID: 38454919 PMCID: PMC10918269 DOI: 10.1016/j.jsps.2024.101999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
The purpose of this study was to enhance the topical delivery of 5-Fluorouracil (5-FU), a cancer treatment, by developing a nanoemulgel formulation. Glycyrrhizin (GLY), a natural penetration enhancer has been investigated to exhibit synergistic effects with 5-FU in inhibiting melanoma cell proliferation and inducing apoptosis, Hence, GLY, along with suitable lipids was utilized to create an optimized nanoemulsion (NE) based gel. Solubility studies and ternary phase diagram revealed isopropyl myristate (IPM), Span 80, Tween 80 as Smix and Transcutol P as co-surfactant. IPM demonstrates excellent solubilizing properties facilitates higher drug loading, ensuring efficient delivery to the target site.,The optimized formulation consisting of 40 % IPM, 30 % of mixture of Tween80: Span80 (Smix) and 15 % Transcutol P provides with a nanometric size of 64.1 ± 5.13 nm and drug loading of 97.3 ± 5.83 %. The optimized formulation observed with no creaming and breakeing of NE and found thermodynamically stable during different stress conditions (temperatures of 4.0 °C and 45.0 °C) and physical thawing (-21.0 ± 0.50 °C to 20.0 ± 0.50 °C). The NE was then transformed into a nanoemulgel (NEG) using 1.5 % w/w Carbopol base and 0.1 % w/w glycyrrhizin. The ex vivo permeability studies showed significant enhancements in drug permeability with the GLY-based 5-FU-NEG formulation compared to pure 5-FU gel in excised pig skin upto1440 min in PBS 7.4 as receptor media. The IC50 values for Plain 5-FU gel, 5-FU-NEG, and GLY-based 5-FU-NEG were found to be 20 µg/mL, 1.1 µg/mL, and 0.1 µg/mL, respectively in B16F10 cell lines. The percentage intracellular uptake of GLY-5-FU-NEG and 5-FU-NEG was found to be 44.3 % and 53.6 %, respectively. GLY-based 5-FU-NEG formulation showed alterations in cell cycle distribution, in compared to 5-FU-NE gel. The overall findings suggest that the GLY-based 5-FU-NEG holds promise for improving anti-melanoma activity.
Collapse
Affiliation(s)
- Nimish Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - G.D. Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Karan Razdan
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Norah A. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Dilpreet Singh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
- University Institute of Pharma Sciences, Chandigarh University, Gharuan (140413), Mohali, India
| |
Collapse
|
7
|
Li D, Wang X, Park DJ, Lee DH, Oh S. Inhibitory Effects of Latilactobacillus curvatus BYB3 Cell-Free Extract on Human Melanoma B16F10 Cells and Tumorigenic Mice. J Microbiol Biotechnol 2024; 34:589-595. [PMID: 38044715 PMCID: PMC11016762 DOI: 10.4014/jmb.2309.09002] [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: 09/04/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 12/05/2023]
Abstract
Latilactobacillus curvatus BYB3 (BYB3) is a species of lactic acid bacteria, formerly named Lactobacillus curvatus, which is isolated from kimchi. In this study, the effect of BYB3, Lactobacillus rhamnosus GG, and Lactobacillus acidophilus GP1B strain extracts at various concentrations was examined on B16F10, a mouse melanoma cell line. Cell viability was examined via MTT assay, and the results indicated that compared to the other two probiotics, BYB3 significantly decreased the total percentages of viable cells. The effects of BYB3 on cell migration and proliferation in B16F10 cells were evaluated using wound healing mobility and proliferation assays, respectively; the results indicated that BYB3 inhibits cell migration and proliferation in a concentration-dependent manner. Using human dermal fibroblast cells to investigate BYB3 extract in vivo had no effect on skin-related cells. Nonetheless, the BYB3 extract inhibited tumor growth in a mouse model, as demonstrated by liver slices. Therefore, this suggests that using BYB3 extract to inhibit melanoma may be a novel approach.
Collapse
Affiliation(s)
- Dingyun Li
- Division of Animal Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Xing Wang
- Division of Animal Science, Chonnam National University, Gwangju 61186, Republic of Korea
- Department of Biochemistry Microbiology and Immunology, Wayne State University, Detroit, MI 48202, USA
| | - Dong-June Park
- Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
| | - Dong Hun Lee
- Department of Biological Sciences, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Sejong Oh
- Division of Animal Science, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
8
|
Dachani S, Kaleem M, Mujtaba MA, Mahajan N, Ali SA, Almutairy AF, Mahmood D, Anwer MK, Ali MD, Kumar S. A Comprehensive Review of Various Therapeutic Strategies for the Management of Skin Cancer. ACS OMEGA 2024; 9:10030-10048. [PMID: 38463249 PMCID: PMC10918819 DOI: 10.1021/acsomega.3c09780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024]
Abstract
Skin cancer (SC) poses a global threat to the healthcare system and is expected to increase significantly over the next two decades if not diagnosed at an early stage. Early diagnosis is crucial for successful treatment, as the disease becomes more challenging to cure as it progresses. However, identifying new drugs, achieving clinical success, and overcoming drug resistance remain significant challenges. To overcome these obstacles and provide effective treatment, it is crucial to understand the causes of skin cancer, how cells grow and divide, factors that affect cell growth, and how drug resistance occurs. In this review, we have explained various therapeutic approaches for SC treatment via ligands, targeted photosensitizers, natural and synthetic drugs for the treatment of SC, an epigenetic approach for management of melanoma, photodynamic therapy, and targeted therapy for BRAF-mutated melanoma. This article also provides a detailed summary of the various natural drugs that are effective in managing melanoma and reducing the occurrence of skin cancer at early stages and focuses on the current status and future prospects of various therapies available for the management of skin cancer.
Collapse
Affiliation(s)
- Sudharshan
Reddy Dachani
- Department
of Pharmacy Practice, College of Pharmacy, Shaqra University, Al-Dawadmi Campus, Al-Dawadmi 11961, Saudi Arabia
| | - Mohammed Kaleem
- Department
of Pharmacology, Babasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440037, Maharashtra, India
| | - Md. Ali Mujtaba
- Department
of Pharmaceutics, Faculty of Pharmacy, Northern
Border University, Arar 91911, Saudi Arabia
| | - Nilesh Mahajan
- Department
of Pharmaceutics, Dabasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440037, Maharashtra, India
| | - Sayyed A. Ali
- Department
of Pharmaceutics, Dabasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440037, Maharashtra, India
| | - Ali F Almutairy
- Department
of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Danish Mahmood
- Department
of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Md. Khalid Anwer
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Mohammad Daud Ali
- Department
of Pharmacy, Mohammed Al-Mana College for
Medical Sciences, Abdulrazaq Bin Hammam Street, Al Safa 34222, Dammam, Saudi Arabia
| | - Sanjay Kumar
- Department
of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Uttar Pradesh 201306, India
| |
Collapse
|
9
|
Adamus-Grabicka AA, Hikisz P, Sikora J. Nanotechnology as a Promising Method in the Treatment of Skin Cancer. Int J Mol Sci 2024; 25:2165. [PMID: 38396841 PMCID: PMC10889690 DOI: 10.3390/ijms25042165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The incidence of skin cancer continues to grow. There are an estimated 1.5 million new cases each year, of which nearly 350,000 are melanoma, which is often fatal. Treatment is challenging and often ineffective, with conventional chemotherapy playing a limited role in this context. These disadvantages can be overcome by the use of nanoparticles and may allow for the early detection and monitoring of neoplastic changes and determining the effectiveness of treatment. This article briefly reviews the present understanding of the characteristics of skin cancers, their epidemiology, and risk factors. It also outlines the possibilities of using nanotechnology, especially nanoparticles, for the transport of medicinal substances. Research over the previous decade on carriers of active substances indicates that drugs can be delivered more accurately to the tumor site, resulting in higher therapeutic efficacy. The article describes the application of liposomes, carbon nanotubes, metal nanoparticles, and polymer nanoparticles in existing therapies. It discusses the challenges encountered in nanoparticle therapy and the possibilities of improving their performance. Undoubtedly, the use of nanoparticles is a promising method that can help in the fight against skin cancer.
Collapse
Affiliation(s)
- Angelika A. Adamus-Grabicka
- Department of Bioinorganic Chemistry, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland;
| | - Pawel Hikisz
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Joanna Sikora
- Department of Bioinorganic Chemistry, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland;
| |
Collapse
|
10
|
Zhang D, Zhang M, Pang Y, Li M, Ma W. Folic Acid-Modified Long-Circulating Liposomes Loaded with Sulfasalazine For Targeted Induction of Ferroptosis in Melanoma. ACS Biomater Sci Eng 2024; 10:588-598. [PMID: 38117929 DOI: 10.1021/acsbiomaterials.3c01223] [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: 12/22/2023]
Abstract
Melanoma is a malignant tumor that originates from melanocytes. The incidence of melanoma is increasing worldwide, partially because of its insensitivity to radiotherapy or chemotherapy. Therefore, effective treatments for melanoma are urgently required. In this study, we employed folic acid-modified sulfasalazine long-circulating liposomes (FA-SSZ-Lips) to precisely target drug delivery to melanoma cells, eliciting ferroptosis effectively. The synthesized FA-SSZ-Lips were characterized as small spheres of a double-layer membrane, a particle size of 110.1 nm, and a ζ-potential of -22.8 ± 0.66 mV. FA-SSZ-Lips are effective drug carriers with SSZ-loading ratio and SSZ release rate of 6.2 ± 0.10%, and 72.63 ± 1.40%, respectively. The liposomes enhanced SSZ solubility, and the folic acid modifications increased the liposome targeting to melanoma cells. Compared with SSZ alone, FA-SSZ-Lips more strongly inhibited B16F10 cell growth, significantly disrupted the intracellular redox balance, and induced ferroptosis. After treatment, considerable differences were observed in the tumor volumes between FA-SSZ-Lips and phosphate-buffered saline control groups. The tumor growth-inhibition value of the FA-SSZ-Lips group reached 70.09%. Thus, FA-SSZ-Lips exhibited favorable antitumor effects in vitro and in vivo and are a promising strategy for melanoma treatment.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Mogen Zhang
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Yunyan Pang
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Meiling Li
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Weiyuan Ma
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| |
Collapse
|
11
|
Dana PM, Hallajzadeh J, Asemi Z, Mansournia MA, Yousefi B. Advances in Chitosan-based Drug Delivery Systems in Melanoma: A Narrative Review. Curr Med Chem 2024; 31:3488-3501. [PMID: 37202890 DOI: 10.2174/0929867330666230518143654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 05/20/2023]
Abstract
Melanoma accounts for the minority of skin cancer cases. However, it has the highest mortality rate among the subtypes of skin cancer. At the early stages of the disease, patients show a good prognosis after the surgery, but developing metastases leads to a remarkable drop in patients' 5-year survival rate. Despite the advances made in the therapeutic approaches to this disease, melanoma treatment is still facing several obstacles. Systemic toxicity, water insolubility, instability, lack of proper biodistribution, inadequate cellular penetration, and rapid clearance are some of the challenges that should be addressed in the field of melanoma treatment. While various delivery systems have been developed to circumvent these challenges, chitosan-based delivery platforms have indicated significant success. Chitosan that is produced by the deacetylation of chitin can be formulated into different materials (e.g., nanoparticle, film, and hydrogel) due to its characteristics. Both in vitro and in vivo studies have reported that chitosan-based materials can be used in drug delivery systems while offering a solution for the common problems in this area, such as enhancing biodistribution and skin penetration as well as the sustained release of the drugs. Herein, we reviewed the studies concerning the role of chitosan as a drug delivery system in melanoma and discussed how these drug systems are used for delivering chemotherapeutic drugs (e.g., doxorubicin and paclitaxel), genes (e.g., TRAIL), and RNAs (e.g., miRNA199a and STAT3 siRNA) successfully. Furthermore, we take a look into the role of chitosan-based nanoparticles in neutron capture therapy.
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Saeidi Z, Giti R, Rostami M, Mohammadi F. Nanotechnology-Based Drug Delivery Systems in the Transdermal Treatment of Melanoma. Adv Pharm Bull 2023; 13:646-662. [PMID: 38022807 PMCID: PMC10676549 DOI: 10.34172/apb.2023.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 01/15/2023] [Accepted: 01/20/2023] [Indexed: 12/01/2023] Open
Abstract
The incidence rate of melanoma is dramatically increasing worldwide, raising it to the fifth most common cancer in men and the sixth in women currently. Resistance generally occurs to the agents used in chemotherapy; besides their high toxicity destroys the normal cells. This study reviewed a detailed summary of the structure, advantages, and disadvantages of nanotechnology-based drug delivery systems in the treatment of melanoma, as well as some nanocarrier applications in animal models or clinical studies. Respective databases were searched for the target keywords and 93 articles were reviewed and discussed. A close study of the liposomes, niosomes, transferosomes, ethosomes, transethosomes, cubosomes, dendrimers, cyclodextrins, solid lipid nanoparticles, and carbon nanotubes (CNTs) was conducted. It was found that these nanocarriers could inhibit metastasis and migration of melanoma cells and decrease cell viability. Conclusively, some nanocarriers like liposomes, niosomes, and transferosomes have been discussed as superior to conventional therapies for melanoma treatment.
Collapse
Affiliation(s)
- Zahra Saeidi
- Department of Pharmaceutics, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Rashin Giti
- Department of Prosthodontics, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Rostami
- Department of Pharmaceutics, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farhad Mohammadi
- Department of Pharmaceutics, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
13
|
Xu M, Li S. Nano-drug delivery system targeting tumor microenvironment: A prospective strategy for melanoma treatment. Cancer Lett 2023; 574:216397. [PMID: 37730105 DOI: 10.1016/j.canlet.2023.216397] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
Melanoma, the most aggressive form of cutaneous malignancy arising from melanocytes, is frequently characterized by metastasis. Despite considerable progress in melanoma therapies, patients with advanced-stage disease often have a poor prognosis due to the limited efficacy, off-target effects, and toxicity associated with conventional drugs. Nanotechnology has emerged as a promising approach to address these challenges with nanoparticles capable of delivering therapeutic agents specifically to the tumor microenvironment (TME). However, the clinical approval of nanomedicines for melanoma treatment remains limited, necessitating further research to develop nanoparticles with improved biocompatibility and precise targeting capabilities. This comprehensive review provides an overview of the current research on nano-drug delivery systems for melanoma treatment, focusing on liposomes, polymeric nanoparticles, and inorganic nanoparticles. It discusses the potential of these nanoparticles for targeted drug delivery, as well as their ability to enhance the efficacy of conventional drugs while minimizing toxicity. Furthermore, this review emphasizes the significance of interdisciplinary collaboration between researchers from various fields to advance the development of nanomedicines. Overall, this review serves as a valuable resource for researchers and clinicians interested in the potential of nano-drug delivery systems for melanoma treatment and offers insights into future directions for research in this field.
Collapse
Affiliation(s)
- Mengdan Xu
- Department of Hematology and Breast Cancer, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, China.
| |
Collapse
|
14
|
Bialves TS, Bastos Junior CLQ, Cordeiro MF, Boyle RT. Snake venom, a potential treatment for melanoma. A systematic review. Int J Biol Macromol 2023; 231:123367. [PMID: 36690229 DOI: 10.1016/j.ijbiomac.2023.123367] [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: 10/11/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Despite advances in treating patients with melanoma, there are still many treatment challenges to overcome. Studies with snake venom-derived proteins/peptides describe their binding potential, and inhibition of some proliferative mechanisms in melanoma. The combined use of these compounds with current therapies could be the strategic gap that will help us discover more effective treatments for melanoma. The present study aimed to carry out a systematic review identifying snake venom proteins and peptides described in the literature with antitumor, antimetastatic, or antiangiogenic effects on melanoma and determine the mechanisms of action that lead to these anti-tumor effects. Snake venoms contain proteins and peptides which are antiaggregant, antimetastatic, and antiangiogenic. The in vivo results are encouraging, considering the reduction of metastases and tumor size after treatment. In addition to these results, it was reported that these venom compounds could act in combination with chemotherapeutics (Acurhagin-C; Macrovipecetin), sensitizing and preparing tumor cells for treatment. There is a consensus that snake venom is a promising strategy for the improvement of antimelanoma therapies, but it has been little explored in the current context, combined with inhibitors, immunotherapy or tumor microenvironment, for example. We suggest Lebein as a candidate for combination therapy with BRAF inhibitors.
Collapse
Affiliation(s)
- Tatiane Senna Bialves
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil.
| | - Claudio L Q Bastos Junior
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil
| | - Marcos Freitas Cordeiro
- Programa de Pós-Graduação em Biociências e Saúde (PPGBS), Universidade do Oeste de Santa Catarina - UNOESC, Rua Roberto Trompovski 224, Joaçaba, Santa Catarina, CEP 89600-000, Brazil.
| | - Robert Tew Boyle
- Programa de Pós-Graduação em Ciências Fisiológicas (PPGCF), Universidade Federal do Rio Grande - FURG, Av. Itália, s/n - km 8 - Carreiros, Rio Grande, Rio Grande do Sul, Brazil; Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, Rio Grande do Sul 96203-900, Brazil
| |
Collapse
|
15
|
Prajapat VM, Mahajan S, Paul PG, Aalhate M, Mehandole A, Madan J, Dua K, Chellappan DK, Singh SK, Singh PK. Nanomedicine: A pragmatic approach for tackling melanoma skin cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
16
|
Pereira I, Monteiro C, Pereira-Silva M, Peixoto D, Nunes C, Reis S, Veiga F, Hamblin MR, Paiva-Santos AC. Nanodelivery systems for cutaneous melanoma treatment. Eur J Pharm Biopharm 2023; 184:214-247. [PMID: 36773725 DOI: 10.1016/j.ejpb.2023.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 10/03/2022] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
Cutaneous melanoma (CM) is a multifactorial disease whose treatment still presents challenges: the rapid progression to advanced CM, which leads to frequent recurrences even after surgical excision and, notably, the low response rates and resistance to the available therapies, particularly in the case of unresectable metastatic CM. Thereby, alternative innovative therapeutic approaches for CM continue to be searched. In this review we discuss relevant preclinical research studies, and provide a broad-brush analysis of patents and clinical trials which involve the application of nanotechnology-based delivery systems in CM therapy. Nanodelivery systems have been developed for the delivery of anticancer biomolecules to CM, which can be administered by different routes. Overall, nanosystems could promote technological advances in several therapeutic modalities and can be used in combinatorial therapies. Nevertheless, the results of these preclinical studies have not been translated to clinical applications. Thus, concerted and collaborative research studies involving basic, applied, translational, and clinical scientists need to be performed to allow the development of effective and safe nanomedicines to treat CM.
Collapse
Affiliation(s)
- Irina Pereira
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Carina Monteiro
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Cláudia Nunes
- LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
17
|
Peng K, Zhang Y, Liu D, Chen J. MMP2 is a immunotherapy related biomarker and correlated with cancer-associated fibroblasts infiltrate in melanoma. Cancer Cell Int 2023; 23:26. [PMID: 36788565 PMCID: PMC9930295 DOI: 10.1186/s12935-023-02862-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/28/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Mounting evidence supports that matrix metalloproteinase (MMPs) are highly associated with tumor progression and that targeting MMPs may overcome the barrier of immune suppression. Among these, whether MMP2 functions as an immunosuppressive role in melanoma, remains unclear. METHODS The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis 2 (GEPIA2) databases were used to assess the prognosis of MMP2 in melanoma, after which Tumor immune estimation resource (TIMER) was used to explore the relationship between MMP2 expression and cancer associated fibroblasts (CAFs) infiltration. Finally, we evaluated the efficacy of MMP2 inhibitor on CAFs infiltration and immunotherapy using a mouse melanoma model. RESULTS In general, the expression of MMP2, MMP13, MMP16, MMP17 and MMP25 were significantly associated with skin cutaneous melanoma (SKCM) patients prognosis, among which MMP2 low expression benefited patients the most. Especially, the overall survival (OS) of BRAF mutation patients with high MMP2 expression was significantly lower than the MMP2 low expression group, but there was no significant difference in BRAF wild-type patients. KEGG and GO enrichment analysis indicated that MMP2 related genes were mostly associated with extracellular structure organization, collagen-containing extracellular matrix and extracellular matrix structural constituent. Furthermore, in almost all cancers, MMP2 expression was positively correlated with CAFs infiltration. MMP2 inhibitor works synergistically with PD-1 antibody and induces tumor regression in a mouse melanoma model, which is dependent on decreased CAFs infiltration. CONCLUSIONS This suggests that MMP2 plays a vital role in the regulation of CAFs infiltration, potentially participating in immunotherapy response, and thus representing a valuable target of immunotherapy in melanoma.
Collapse
Affiliation(s)
- Kunwei Peng
- grid.412534.5Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Medical Oncology, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Guangzhou, 510260 Guangdong People’s Republic of China
| | - Yanyan Zhang
- grid.79703.3a0000 0004 1764 3838Department of Infectious Diseases, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong People’s Republic of China
| | - Deyi Liu
- grid.412534.5Department of General Practice, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong People’s Republic of China
| | - Jingqi Chen
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Medical Oncology, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Guangzhou, 510260, Guangdong, People's Republic of China. .,Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
18
|
Fu QT, Zhong XQ, Chen MY, Gu JY, Zhao J, Yu DH, Tan F. Luteolin-Loaded Nanoparticles for the Treatment of Melanoma. Int J Nanomedicine 2023; 18:2053-2068. [PMID: 37101838 PMCID: PMC10124627 DOI: 10.2147/ijn.s400329] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/29/2023] [Indexed: 04/28/2023] Open
Abstract
Background and Purpose Luteolin (LUT), a flavonoid found in various plants, has been reported to have potential therapeutic effects in melanoma. However, poor water solubility and low bioactivity have severely restricted the clinical application of LUT. Based on the high reactive oxygen species (ROS) levels in melanoma cells, we developed nanoparticles encapsulating LUT with the ROS-responsive material poly(propylene sulfide)-poly(ethylene glycol) (PPS-PEG) to enhance the water solubility of LUT, accelerate the release of LUT in melanoma cells, and further enhance its anti-melanoma effect, providing a viable solution for the application of LUT nano-delivery systems in melanoma therapy. Methods In this study, LUT-loaded nanoparticles were prepared with PPS-PEG and named as LUT-PPS-NPs. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) were applied to determine the size and morphology of LUT-PPS-NPs. In vitro studies were carried out to determine the uptake and mechanism of LUT-PPS-NPs by SK-MEL-28 melanoma cells. According to the CCK-8 assay, the cytotoxic effects of LUT-PPS-NPs on human skin fibroblasts (HSF) and SK-MEL-28 cells were assessed. Apoptosis assays, cell migration and invasion assays, and proliferation inhibition assays with low and normal density plating were also applied to test the in vitro anti-melanoma effect. Additionally, melanoma models were established utilizing BALB/c nude mice and initially evaluated the growth inhibitory impact following intratumoral injection of LUT-PPS-NPs. Results The size of LUT-PPS-NPs was 169.77 ± 7.33 nm with high drug loading (15.05 ± 0.07%). In vitro, cellular assays confirmed that LUT-PPS-NPs were efficiently internalized by SK-MEL-28 cells and showed low cytotoxicity against HSF. Moreover, LUT released from LUT-PPS-NPs significantly inhibited tumor cell proliferation, migration and invasion. Animal experiments showed that LUT-PPS-NPs inhibited tumor growth more than 2-fold compared with the LUT group. Conclusion In conclusion, the LUT-PPS-NPs developed in our study enhanced the anti-melanoma effect of LUT.
Collapse
Affiliation(s)
- Qiao-Ting Fu
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai, 200443, People’s Republic of China
| | - Xiao-Qin Zhong
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai, 200443, People’s Republic of China
| | - Mei-Yu Chen
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai, 200443, People’s Republic of China
| | - Jia-Yi Gu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, People’s Republic of China
| | - Jian Zhao
- Department of Oncology-Pathology, Karolinska Institutet, BioClinicum, Karolinska University Hospital Solna, Solna, Sweden
| | - De-Hong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, People’s Republic of China
| | - Fei Tan
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai, 200443, People’s Republic of China
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Correspondence: Fei Tan; Dehong Yu, Email ;
| |
Collapse
|
19
|
Jia F, Yu W, Li X, Chen Y, Wang Y, Ji J. Microneedles loaded with glutathione-scavenging composites for nitric oxide enhanced photodynamic therapy of melanoma. Bioeng Transl Med 2023; 8:e10352. [PMID: 36684091 PMCID: PMC9842046 DOI: 10.1002/btm2.10352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/26/2022] [Accepted: 05/13/2022] [Indexed: 01/25/2023] Open
Abstract
Photodynamic therapy (PDT) represents an attractive promising route for melanoma treatment. However, its therapeutic efficacy is compromised by inefficient drug delivery and high glutathione (GSH) levels in cancer cells. To overcome these challenges, microneedles (MNs) system loaded with GSH-scavenging nanocomposites was presented for nitric oxide (NO) enhanced PDT. The nanocomposites consisted of S-nitroso-N-acrylate penicillamine (SNAP; a NO donor) grafted fourth-generation polyamide amine dendrimer (G4) and chlorin e6 (Ce6). Upon local insertion of polyvinylpyrrolidone MNs, G4-SNAP/Ce6 composites were fast delivered and significantly amplified the therapeutic effects during PDT, via GSH depletion and reactive nitrogen species generation. Even with a single administration and low power light exposure, MNs with G4-SNAP/Ce6 effectively halt the tumor progression. The system demonstrated better cancer ablation efficacy than Ce6 alone toward melanoma. The strategy may inspire new ideas for future PDT-related therapy for skin tumors.
Collapse
Affiliation(s)
- Fan Jia
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
| | - Weijiang Yu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
| | - Xinfang Li
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
| | - Yonghang Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
20
|
Viegas JSR, Bentley MVLB, Vicentini FTMDC. Challenges to perform an efficiently gene therapy adopting non-viral vectors: Melanoma landscape. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
21
|
Jamshidi-Adegani F, Vakilian S, Al-Hashmi S, Al-Kindi J, Rehman NU, Al-Sinani Y, Ghaemi S, Alam K, Anwar MU, Csuk R, Al-Harrasi A. Selective anti-cancer activity against melanoma cells using 3- O-acetyl-β-boswellic acid-loaded 3D-Printed scaffold. Nat Prod Res 2022; 37:2049-2054. [PMID: 36008779 DOI: 10.1080/14786419.2022.2116024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
This study aimed to develop a local 3 D-printed bioactive graft using poly-caprolacton (PCL) as a drug carrier and 3-O-acetyl-β-boswellic acid (β-ABA) as an anticancer compound. β-ABA-loaded 3 D-printed scaffold was fabricated and physically characterized. The results indicated more desirable mechanical and physical properties of the β-ABA-loaded PCL mat in comparison with the PCL scaffold. Following sustained release of β-ABA, the β-ABA-loaded PCL scaffold revealed selective cytotoxic activity against melanoma cells, while the PCL + ABA with the bolus delivery of β-ABA was toxic against fibroblast cells. Followed by the induction of apoptosis in melanoma cells at the gene level, the result of the western blot showed that the β-ABA-loaded scaffold significantly up-regulated P53 and down-regulated BCL2, with an increment in the ratio of Bax/BCL2. The selective anti-cancer properties of β-ABA-loaded 3 D printed scaffold against melanoma cells indicated that this scaffold could be potentially used as a bioactive graft to improve the melanoma treatment.
Collapse
Affiliation(s)
- Fatemeh Jamshidi-Adegani
- Laboratory for Stem Cell & Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Saeid Vakilian
- Laboratory for Stem Cell & Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Sulaiman Al-Hashmi
- Laboratory for Stem Cell & Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Juhaina Al-Kindi
- Laboratory for Stem Cell & Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Najeeb Ur Rehman
- Natural products Laboratory, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Yaqeen Al-Sinani
- Laboratory for Stem Cell & Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Shokoofeh Ghaemi
- Laboratory for Stem Cell & Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman.,Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Khurshid Alam
- Department of Mechanical and Industrial Engineering, Sultan Qaboos University, Muscat, Oman
| | - Muhammad U Anwar
- X-Ray Diffraction & Crystallography Lab, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, PC, Oman
| | - Rene Csuk
- Organic Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Ahmed Al-Harrasi
- Natural products Laboratory, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| |
Collapse
|
22
|
Tian C, Zheng S, Liu X, Kamei KI. Tumor-on-a-chip model for advancement of anti-cancer nano drug delivery system. J Nanobiotechnology 2022; 20:338. [PMID: 35858898 PMCID: PMC9301849 DOI: 10.1186/s12951-022-01552-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/12/2022] [Indexed: 12/27/2022] Open
Abstract
Despite explosive growth in the development of nano-drug delivery systems (NDDS) targeting tumors in the last few decades, clinical translation rates are low owing to the lack of efficient models for evaluating and predicting responses. Microfluidics-based tumor-on-a-chip (TOC) systems provide a promising approach to address these challenges. The integrated engineered platforms can recapitulate complex in vivo tumor features at a microscale level, such as the tumor microenvironment, three-dimensional tissue structure, and dynamic culture conditions, thus improving the correlation between results derived from preclinical and clinical trials in evaluating anticancer nanomedicines. The specific focus of this review is to describe recent advances in TOCs for the evaluation of nanomedicine, categorized into six sections based on the drug delivery process: circulation behavior after infusion, endothelial and matrix barriers, tumor uptake, therapeutic efficacy, safety, and resistance. We also discuss current issues and future directions for an end-use perspective of TOCs.
Collapse
Affiliation(s)
- Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China.,Chinese People's Liberation Army 210 Hospital, 116021, Dalian, People's Republic of China
| | - Shunzhe Zheng
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Xinying Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China
| | - Ken-Ichiro Kamei
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, People's Republic of China. .,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, 606-8501, Kyoto, Japan.
| |
Collapse
|
23
|
Sun VZ, Melim TL, Mitra S, Erickson JE, Bryant SH, Farnham A, Westmoreland S, Knight H, Zhang L, Ritacco W, Homan K, Benatuil L, Sterman AJS, Goodearl AD. Fibronectin extra domain A as a drug delivery targeting epitope for rheumatoid arthritis. Adv Rheumatol 2022; 62:17. [PMID: 35624488 DOI: 10.1186/s42358-022-00247-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 05/15/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES To assess the ability of monoclonal antibodies (mAbs) specific for fibronectin extra-domain A (FnEDA) to target diseased tissues of mouse collagen induced arthritis (mCIA) models. To explore the parameters of the targeting exhibited by anti-FnEDA mAbs including timing and location. METHODS Targeting capabilities of anti-FnEDA mAbs were demonstrated by biodistribution study where i.v. injected antibodies were detected by conjugated near-infrared (NIR) fluorophore, 125I label and immunohistochemistry (IHC) of the injected antibody. Location of FnEDA expression in both mCIA and human RA tissue were mapped by IHC. Quantification of anti-FnEDA mAbs targeted to disease tissue was measured by whole-body autoradiography (WBA). Timing of the targeting was interrogated with fluorescent and confocal microscopy using anti-FnEDA mAbs labeled with different fluorophores and injected at different times. RESULTS Anti-FnEDA mAbs show specific targeting to diseased paws of mCIA animal. The targeting was focused on inflamed synovium which is consistent with FnEDA expression profile in both mCIA and human RA tissues. Anti-FnEDA mAbs accumulated in diseased tissue at pharmacologically relevant concentrations, the targeting was sustained for up to 14 days and FnEDA was able to support targeting of multiple doses of anti-FnEDA mAbs given 5 days apart. CONCLUSION FnEDA is specifically upregulated in the inflamed tissues of mCIA. Antibodies specific for FnEDA can be useful as molecular delivery vehicles for disease specific targeting of payloads to inflamed joint tissue.
Collapse
Affiliation(s)
- Victor Z Sun
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA.
| | - Terry L Melim
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Soumya Mitra
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Jamie E Erickson
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Shaughn H Bryant
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| | | | - Susan Westmoreland
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Heather Knight
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Liang Zhang
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| | - Wendy Ritacco
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| | | | | | | | - Andrew D Goodearl
- Drug Discovery Science and Technology, Abbvie Bioresearch Center, Worcester, MA, USA
| |
Collapse
|
24
|
Overview of Application of Nanomaterials in Medical Domain. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:3507383. [PMID: 35601569 PMCID: PMC9095353 DOI: 10.1155/2022/3507383] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 01/13/2022] [Indexed: 11/19/2022]
Abstract
With the development of nanotechnology, the application of nanomaterials in the medical field has become a forefront hotspot in the field of scientific research in the 21st century. Compared with traditional drug carriers, drug carriers made of nanomaterials have advantages such as higher drug loading rate, better biocompatibility, and targeted transportation, which provide the possibility for the treatment of a variety of diseases. In this paper, the characteristics and advantages of nanomaterials as well as their applications in the medical field are reviewed and the research progress of nanomaterials is analyzed.
Collapse
|
25
|
Tong S, Darwish S, Ariani HHN, Lozada KA, Salehi D, Cinelli MA, Silverman RB, Kaur K, Yang S. A Small Peptide Increases Drug Delivery in Human Melanoma Cells. Pharmaceutics 2022; 14:1036. [PMID: 35631623 PMCID: PMC9145755 DOI: 10.3390/pharmaceutics14051036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
Melanoma is the most fatal type of skin cancer and is notoriously resistant to chemotherapies. The response of melanoma to current treatments is difficult to predict. To combat these challenges, in this study, we utilize a small peptide to increase drug delivery to melanoma cells. A peptide library array was designed and screened using a peptide array-whole cell binding assay, which identified KK-11 as a novel human melanoma-targeting peptide. The peptide and its D-amino acid substituted analogue (VPWxEPAYQrFL or D-aa KK-11) were synthesized via a solid-phase strategy. Further studies using FITC-labeled KK-11 demonstrated dose-dependent uptake in human melanoma cells. D-aa KK-11 significantly increased the stability of the peptide, with 45.3% remaining detectable after 24 h with human serum incubation. Co-treatment of KK-11 with doxorubicin was found to significantly enhance the cytotoxicity of doxorubicin compared to doxorubicin alone, or sequential KK-11 and doxorubicin treatment. In vivo and ex vivo imaging revealed that D-aa KK-11 distributed to xenografted A375 melanoma tumors as early as 5 min and persisted up to 24 h post tail vein injection. When co-administered, D-aa KK-11 significantly enhanced the anti-tumor activity of a novel nNOS inhibitor (MAC-3-190) in an A375 human melanoma xenograft mouse model compared to MAC-3-190 treatment alone. No apparent systemic toxicities were observed. Taken together, these results suggest that KK-11 may be a promising human melanoma-targeted delivery vector for anti-melanoma cargo.
Collapse
Affiliation(s)
- Shirley Tong
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.T.); (K.A.L.)
| | - Shaban Darwish
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.D.); (H.H.N.A.); (D.S.)
| | - Hanieh Hossein Nejad Ariani
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.D.); (H.H.N.A.); (D.S.)
| | - Kate Alison Lozada
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.T.); (K.A.L.)
| | - David Salehi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.D.); (H.H.N.A.); (D.S.)
| | - Maris A. Cinelli
- Center for Developmental Therapeutics, Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; (M.A.C.); (R.B.S.)
| | - Richard B. Silverman
- Center for Developmental Therapeutics, Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; (M.A.C.); (R.B.S.)
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kamaljit Kaur
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.D.); (H.H.N.A.); (D.S.)
| | - Sun Yang
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA 92618, USA; (S.T.); (K.A.L.)
| |
Collapse
|
26
|
Li M, Zhao J, Yang R, Cai R, Liu X, Xie J, Shu B, Qi S. CENPF as an independent prognostic and metastasis biomarker corresponding to CD4+ memory T cells in cutaneous melanoma. Cancer Sci 2022; 113:1220-1234. [PMID: 35189004 PMCID: PMC8990861 DOI: 10.1111/cas.15303] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 12/01/2022] Open
Abstract
Owing to recent advances in immunotherapies, the overall survival of patients with skin cutaneous melanoma (SKCM) has increased; however, the 5-year survival rate of metastatic patients remains poor. Skin cutaneous melanoma-upregulated genes were screened via analysis of differentially expressed genes (GSE3189 and GSE46517), and metastasis-related oncogenes were identified via weighted gene coexpression network analysis of the GSE46517 dataset. As confirmed by the Tumor Immune Estimation Resource, we found highly expressed centromere protein F (CENPF) in SKCM and its metastases. Immunostaining of human melanoma tissues demonstrated high CENPF expression. According to the Kaplan-Meier survival curve log-rank test, receiver-operating characteristic curve, and univariate and multivariate analyses, the Cancer Genome Atlas (TCGA) database suggested CENPF be a typical independent predictor of SKCM. The CIBERSORT algorithm classified the types of the immune cells from GSE46517 and showed higher proportion of CD4+ memory-activated T cells in metastatic melanoma. Single-sample gene set enrichment analysis of TCGA data confirmed the correlation between CENPF and activated CD4+ T cells. Centromere protein F was positively correlated with tumor mutational burden and CD4+ memory T cell markers (interleukin [IL]-23A, CD28, and CD62L), negatively associated with memory T cell maintenance factors (IL-7 and IL-15) by correlation analysis. Moreover, immunofluorescence showed high coexpression of CENPF and IL23A, CD4 in melanoma. Upregulated CENPF might lead to premature depletion of CD4+ memory T cells and immunosuppression. Nomogram indicated CENPF clinical predictive value for 1-, 3-, 5-, and 7-year melanoma overall survival. Therefore, CENPF plays a vital role in the progression and metastasis of melanoma and can be an effective therapeutic target.
Collapse
Affiliation(s)
- Mengzhi Li
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Jingling Zhao
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Ronghua Yang
- Department of Burn SurgeryThe First People’s Hospital of FoshanFoshanChina
| | - Ruizhao Cai
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xusheng Liu
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Julin Xie
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Bin Shu
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Shaohai Qi
- Department of BurnsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
27
|
Zeng H, Li J, Hou K, Wu Y, Chen H, Ning Z. Melanoma and Nanotechnology-Based Treatment. Front Oncol 2022; 12:858185. [PMID: 35356202 PMCID: PMC8959641 DOI: 10.3389/fonc.2022.858185] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Melanoma is a malignant tumor arising in melanocytes from the basal layer of the epidermis and is the fifth most commonly diagnosed cancer in the United States. Melanoma is aggressive and easily metastasizes, and the survival rate is low. Nanotechnology-based diagnosis and treatment of melanoma have attracted increasing attention. Importantly, nano drug delivery systems have the advantages of increasing drug solubility, enhancing drug stability, prolonging half-life, optimizing bioavailability, targeting tumors, and minimizing side effects; thus, these systems can facilitate tumor cytotoxicity to achieve effective treatment of melanoma. In this review, we discuss current nanosystems used in the diagnosis and treatment of melanoma, including lipid systems, inorganic nanoparticles, polymeric systems, and natural nanosystems. The excellent characteristics of novel and effective drug delivery systems provide a basis for the broad applications of these systems in the diagnosis and treatment of melanoma, particularly metastatic melanoma.
Collapse
Affiliation(s)
- Hong Zeng
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Li
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Hou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo Chen
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeng Ning
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
3, 3'- (3, 5-DCPBC) Down-Regulates Multiple Phosphokinase Dependent Signal Transduction Pathways in Malignant Melanoma Cells through Specific Diminution of EGFR Y1086 Phosphorylation. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041172. [PMID: 35208960 PMCID: PMC8874408 DOI: 10.3390/molecules27041172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/19/2022]
Abstract
Melanoma is the most dangerous skin malignancy due to its strong metastatic potential with high mortality. Activation of crucial signaling pathways enforcing melanoma progression depends on phosphorylation of distinct tyrosine kinases and oxidative stress. We here investigated the effect of a bis-coumarin derivative [3, 3′- ((3″, 5′-Dichlorophenyl) methylene) bis (4-hydroxy-2H-chromen-2-one)] [3, 3′- (3, 5-DCPBC)] on human melanoma cell survival, growth, proliferation, migration, intracellular redox state, and deciphered associated signaling pathways. This derivative is toxic for melanoma cells and non-toxic for melanocytes, their benign counterpart, and fibroblasts. 3, 3′- (3, 5-DCPBC) inhibits cell survival, migration, and proliferation of different metastatic and non-metastatic melanoma cell lines through profound suppression of the phosphorylation of Epidermal Growth Factor receptor (EGFR) and proto-oncogene cellular sarcoma (c-SRC) related downstream pathways. Thus, 3, 3′- (3, 5-DCPBC) endowed with the unique property to simultaneously suppress phosphorylation of multiple downstream kinases, such as EGFR/JAK/STAT and EGFR/SRC and their corresponding transcription factors.
Collapse
|
29
|
NEAT1 in bone marrow mesenchymal stem cell-derived extracellular vesicles promotes melanoma by inducing M2 macrophage polarization. Cancer Gene Ther 2022; 29:1228-1239. [PMID: 35115683 DOI: 10.1038/s41417-021-00392-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/23/2021] [Accepted: 09/22/2021] [Indexed: 12/25/2022]
Abstract
Bone marrow mesenchymal stem cells (BMSCs)-derived extracellular vesicles (EVs) reportedly play an important role in melanoma pathogenesis. This study aimed to explore the mechanisms of EVs-carried long non-coding RNA (lncRNA) NEAT1 involvement in melanoma. Gain- and loss-of-function experiments were performed to determine biological characteristics of A-375 melanoma cells. Bioinfomatic prediction, RNA immunoprecipitation (RIP), and dual luciferase reporter gene experiments were applied to investigate the roles of NEAT1 and microRNA-374a-5p (miR-374a-5p), and leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4). A subcutaneous tumor model was constructed using nude mice, and in vivo fluorescence imaging was used to observe the effect of NEAT1 on the growth and metastasis of melanoma cells in vivo. The results indicated that BMSC-EVs could be internalized by macrophages to promote the expression of macrophages M2 markers. M2 type macrophages promoted malignancy of melanoma cells. NEAT1 derived from BMSC-EVs promoted the progression of melanoma by promoting M2 polarization of macrophages. NEAT1 inhibits miR-374 expression, while miR-374 could upregulate LGR4-dependent IQGAP1 expression. The tumor-inhibiting effect of NEAT1 silencing was validated in the nude mouse xenograft model. Collectively, the results demonstrated that BMSC-EVs carrying NEAT1 can promote the progression of melanoma by inducing M2 polarization of macrophages, and thus may be considered as a potential target for melanoma therapeutics.
Collapse
|
30
|
Wei S, Shao X, Liu Y, Xiong B, Cui P, Liu Z, Li Q. Genome editing of PD-L1 mediated by nucleobase-modified polyamidoamine for cancer immunotherapy. J Mater Chem B 2022; 10:1291-1300. [DOI: 10.1039/d1tb02688g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Immune checkpoint blockade therapy against programmed death protein-1 and its ligand (PD-1/PD-L1) has been accepted as a promising approach to activate the immune system's anti-tumor response. Although small interfering RNA...
Collapse
|
31
|
Sonker M, Bajpai S, Khan MA, Yu X, Tiwary SK, Shreyash N. Review of Recent Advances and Their Improvement in the Effectiveness of Hydrogel-Based Targeted Drug Delivery: A Hope for Treating Cancer. ACS APPLIED BIO MATERIALS 2021; 4:8080-8109. [PMID: 35005919 DOI: 10.1021/acsabm.1c00857] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Using hydrogels for delivering cancer therapeutics is advantageous in pharmaceutical usage as they have an edge over traditional delivery, which is tainted due to the risk of toxicity that it imbues. Hydrogel usage leads to the development of a more controlled drug release system owing to its amenability for structural metamorphosis, its higher porosity to seat the drug molecules, and its ability to shield the drug from denaturation. The thing that makes its utility even more enhanced is that they make themselves more recognizable to the body tissues and hence can stay inside the body for a longer time, enhancing the efficiency of the delivery, which otherwise is negatively affected since the drug is identified by the human immunity as a foreign substance, and thus, an attack of the immunity begins on the drug injected. A variety of hydrogels such as thermosensitive, pH-sensitive, and magnetism-responsive hydrogels have been included and their potential usage in drug delivery has been discussed in this review that aims to present recent studies on hydrogels that respond to alterations under a variety of circumstances in "reducing" situations that mimic the microenvironment of cancerous cells.
Collapse
Affiliation(s)
- Muskan Sonker
- Department of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30318, United States
| | - Sushant Bajpai
- Department of Petroleum Engineering, Rajiv Gandhi Institute of Petroleum Technology, Jais, Amethi 229304, India
| | - Mohd Ashhar Khan
- Department of Chemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Jais, Amethi 229304, India
| | - Xiaojun Yu
- Department of Biomedical Engineering Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Saurabh Kr Tiwary
- Department of Chemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Jais, Amethi 229304, India
| | - Nehil Shreyash
- Department of Chemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Jais, Amethi 229304, India
| |
Collapse
|
32
|
Liang S, Wang M, Wang J, Chen G. Red-Blood-Cell-Membrane-Coated Metal-Drug Nanoparticles for Enhanced Chemotherapy. Chembiochem 2021; 22:3184-3189. [PMID: 34468067 DOI: 10.1002/cbic.202100313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/12/2021] [Indexed: 12/11/2022]
Abstract
To overcome high toxicity, low bioavailability and poor water solubility of chemotherapeutics, a variety of drug carriers have been designed. However, most carriers are severely limited by low drug loading capacity and adverse side effects. Here, a new type of metal-drug nanoparticles (MDNs) was designed and synthesized. The MDNs self-assembled with Fe(III) ions and drug molecules through coordination, resulting in nanoparticles with high drug loading. To assist systemic delivery and prolong circulation time, the obtained MDNs were camouflaged with red blood cell (RBCs) membranes (RBCs@Fe-DOX MDNs) to improve their stability and dispersity. The RBCs@Fe-DOX MDNs presented pH-responsive release functionalities, resulting in drug release accelerated in acidic tumor microenvironments. The outstanding in vitro and in vivo antitumor therapeutic outcome was realized by RBCs@Fe-DOX MDNs. This study provides an innovative design guideline for chemotherapy and demonstrates the great capacity of nanomaterials in anticancer treatments.
Collapse
Affiliation(s)
- Shuya Liang
- Department of Dermatology, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Street, Qingdao, Shandong, 266555, P. R. China
| | - Miaomiao Wang
- Department of Dermatology, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Street, Qingdao, Shandong, 266555, P. R. China
| | - Jun Wang
- Department of Dermatology, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Street, Qingdao, Shandong, 266555, P. R. China
| | - Guanzhi Chen
- Department of Dermatology, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Street, Qingdao, Shandong, 266555, P. R. China
| |
Collapse
|
33
|
Demartis S, Rassu G, Murgia S, Casula L, Giunchedi P, Gavini E. Improving Dermal Delivery of Rose Bengal by Deformable Lipid Nanovesicles for Topical Treatment of Melanoma. Mol Pharm 2021; 18:4046-4057. [PMID: 34554752 PMCID: PMC8564756 DOI: 10.1021/acs.molpharmaceut.1c00468] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Cutaneous melanoma
is one of the most aggressive and metastatic
forms of skin cancer. However, current therapeutic options present
several limitations, and the annual death rate due to melanoma increases
every year. Dermal delivery of nanomedicines can effectively eradicate
primary melanoma lesions, avoid the metastatic process, and improve
survival. Rose Bengal (RB) is a sono-photosensitizer drug with intrinsic
cytotoxicity toward melanoma without external stimuli but the biopharmaceutical
profile limits its clinical use. Here, we propose deformable lipid
nanovesicles, also known as transfersomes (TF), for the targeted dermal
delivery of RB to melanoma lesions to eradicate them in the absence
of external stimuli. Considering RB’s poor ability to cross
the stratum corneum and its photosensitizer nature, transfersomal
carriers were selected simultaneously to enhance RB penetration to
the deepest skin layers and protect RB from undesired photodegradation.
RB-loaded TF dispersion (RB-TF), prepared by a modified reverse-phase
evaporation method, were nanosized with a ζ-potential value
below −30 mV. The spectrophotometric and fluorimetric analysis
revealed that RB efficiently interacted with the lipid phase. The
morphological investigations (transmission electron microscopy and
small-angle X-ray scattering) proved that RB intercalated within the
phospholipid bilayer of TF originating unilamellar and deformable
vesicles, in contrast to the rigid multilamellar unloaded ones. Such
outcomes agree with the results of the in vitro permeation study,
where the lack of a burst RB permeation peak for RB-TF, observed instead
for the free drug, suggests that a significant amount of RB interacted
with lipid nanovesicles. Also, RB-TF proved to protect RB from undesired
photodegradation over 24 h of direct light exposure. The ex vivo epidermis
permeation study proved that RB-TF significantly increased RB’s
amount permeating the epidermis compared to the free drug (78.31 vs
38.31%). Finally, the antiproliferative assays on melanoma cells suggested
that RB-TF effectively reduced cell growth compared to free RB at
the concentrations tested (25 and 50 μM). RB-TF could potentially
increase selectivity toward cancer cells. Considering the outcomes
of the characterization and cytotoxicity studies performed on RB-TF,
we conclude that RB-TF represents a valid potential alternative tool
to fight against primary melanoma lesions via dermal delivery in the
absence of light.
Collapse
Affiliation(s)
- Sara Demartis
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Giovanna Rassu
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Sergio Murgia
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Cagliari, Italy.,CSGI, Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Sesto Fiorentino, Florence, Italy
| | - Luca Casula
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Paolo Giunchedi
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Elisabetta Gavini
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
34
|
Kaur H, Kesharwani P. Advanced nanomedicine approaches applied for treatment of skin carcinoma. J Control Release 2021; 337:589-611. [PMID: 34364919 DOI: 10.1016/j.jconrel.2021.08.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022]
Abstract
Skin-cancer is the commonest malignancy affecting huge proportion of the population, reaching heights in terms of morbidity. The treatment strategies are presently focusing on surgery, radiation and chemotherapy, which eventually cause destruction to unaffected cells. To overcome this limitation, wide range of nanoscaled materials have been recognized as potential carriers for delivering selective response to cancerous cells and neoplasms. Nanotechnological approach has been tremendously exploited in several areas, owing to their functional nanometric dimensions. The alarming incidence of skin cancer engenders burdensome effects worldwide, which is further awakening innovational medicinal approaches, accompanying target specific drug delivery tools for coveted benefits to provide reduced toxicity and tackle proliferative episodes of skin cancer. The developed nanosystems for anti-cancer agents include liposomes, ethosomes, nanofibers, solid lipid nanoparticles and metallic nanoparticles, which exhibit pronounced outcomes for skin carcinoma. In this review, skin cancer with its sub-types is explained in nutshell, followed by compendium of specific nanotechnological tools presented, in addition to therapeutic applications of drug-loaded nano systems for skin cancer.
Collapse
Affiliation(s)
- Harsimran Kaur
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
35
|
Feuser PE, Coelho ALS, de Melo ME, Scussel R, Carciofi BAM, Machado-de-Ávila RA, de Oliveira D, de Andrade CJ. Apoptosis Induction in Murine Melanoma (B16F10) Cells by Mannosylerythritol Lipids-B; a Glycolipid Biosurfactant with Antitumoral Activities. Appl Biochem Biotechnol 2021; 193:3855-3866. [PMID: 34302592 DOI: 10.1007/s12010-021-03620-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 07/12/2021] [Indexed: 11/26/2022]
Abstract
Mannosylerythritol lipids have drawn attention to cosmetic and pharmaceutical industries due to their non-toxicity and excellent biological interactions with human skin, particularly with the deepest epidermal layer. Lamellar liquid crystal structure, formed by MEL-B, is an interesting feature due to its similarity to the stratum corneum molecular arrangement and cell signaling events involved in the deregulation of the cancerous cell membrane. Thus, this work aimed to evaluate the cytotoxicity of commercial mannosylerythritol lipids-B in murine melanoma, fibroblast, and human erythrocytes cells. Cytotoxic effect was more pronounced on the tumor cells from 20 µg/mL, reducing cell viability by 65%, whereas fibroblast and human erythrocytes cells were more resistant to glycolipid treatment. Fluorescence microscopy and flow cytometer proved that mannosylerythritol lipids-B is an apoptosis inducer in tumor cells related to reactive oxygen species generation.
Collapse
Affiliation(s)
- P E Feuser
- Graduate Program in Chemical Engineering, Department of Chemical and Food Engineering, Federal University of Santa Catarina, Trindade, s/n, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - A L S Coelho
- Graduate Program in Food Engineering, Department of Chemical and Food Engineering, Federal University of Santa Catarina, Trindade, s/n, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - M E de Melo
- Graduate Program in Health Science, University of the Extreme South Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - R Scussel
- Graduate Program in Health Science, University of the Extreme South Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - B A M Carciofi
- Graduate Program in Food Engineering, Department of Chemical and Food Engineering, Federal University of Santa Catarina, Trindade, s/n, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - R A Machado-de-Ávila
- Graduate Program in Health Science, University of the Extreme South Santa Catarina, Criciúma, Santa Catarina, Brazil
| | - D de Oliveira
- Graduate Program in Chemical Engineering, Department of Chemical and Food Engineering, Federal University of Santa Catarina, Trindade, s/n, Florianópolis, Santa Catarina, 88040-900, Brazil
- Graduate Program in Food Engineering, Department of Chemical and Food Engineering, Federal University of Santa Catarina, Trindade, s/n, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - C J de Andrade
- Graduate Program in Chemical Engineering, Department of Chemical and Food Engineering, Federal University of Santa Catarina, Trindade, s/n, Florianópolis, Santa Catarina, 88040-900, Brazil.
| |
Collapse
|
36
|
Mekkawy AI, Naguib YW, Alhaj-Suliman SO, Wafa EI, Ebeid K, Acri T, Salem AK. Paclitaxel anticancer activity is enhanced by the MEK 1/2 inhibitor PD98059 in vitro and by PD98059-loaded nanoparticles in BRAF V600E melanoma-bearing mice. Int J Pharm 2021; 606:120876. [PMID: 34252520 DOI: 10.1016/j.ijpharm.2021.120876] [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: 04/17/2021] [Revised: 06/24/2021] [Accepted: 07/07/2021] [Indexed: 02/02/2023]
Abstract
Melanoma, the most malignant form of skin cancer, shows resistance to traditional anticancer drugs including paclitaxel (PTX). Furthermore, over 50% of melanoma cases express the BRAFV600E mutation which activates the MAPK pathway increasing cell proliferation and survival. In the current study, we investigated the capacity of the combination therapy of PTX and the MAPK inhibitor, PD98059, to enhance the cytotoxicity of PTX against melanoma and therefore improve treatment outcomes. Synergistic in vitro cytotoxicity was observed when soluble PTX and PD98059 were used to treat the A375 melanoma cell line as evidenced by a significant reduction in the cell viability and IC50 value for PTX. Then, in further studies, TPGS-emulsified PD98059-loaded PLGA nanoparticles (NPs) were prepared, characterized in vitro and assessed for therapeutic efficacy when used in combination with soluble PTX. The average particle size (180 nm d.), zeta potential (-34.8 mV), polydispersity index (0.081), encapsulation efficiency (20%), particle yield (90.8%), and drug loading (6.633 µg/mg) of the prepared NPs were evaluated. Also, cellular uptake and in vitro cytotoxicity studies were performed with these PD98059-loaded NPs and compared to soluble PD98059. The PD98059-loaded NPs were superior to soluble PD98059 in terms of both cellular uptake and in vitro cytotoxicity in A375 cells. In in vivo studies, using A375 challenged mice, we report improved survival in mice treated with soluble PTX and PD98059-loaded NPs. Our findings suggest the potential for using this combinatorial therapy in the management of patients with metastatic melanoma harboring the BRAF mutation as a means to improve survival outcomes.
Collapse
Affiliation(s)
- Aml I Mekkawy
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Sohag University, Sohag, Sohag 82524, Egypt
| | - Youssef W Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Minia 61519, Egypt; Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Manufacturing, Deraya University, New Minia City, Minia 61768, Egypt
| | - Suhaila O Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Kareem Ebeid
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Minia 61519, Egypt; Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Manufacturing, Deraya University, New Minia City, Minia 61768, Egypt
| | - Timothy Acri
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
37
|
Zhi D, Yang T, Zhang T, Yang M, Zhang S, Donnelly RF. Microneedles for gene and drug delivery in skin cancer therapy. J Control Release 2021; 335:158-177. [DOI: 10.1016/j.jconrel.2021.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 12/14/2022]
|
38
|
Liao Y, Jia X, Ren Y, Deji Z, Gesang Y, Ning N, Feng H, Yu H, Wei A. Suppressive role of microRNA-130b-3p in ferroptosis in melanoma cells correlates with DKK1 inhibition and Nrf2-HO-1 pathway activation. Hum Cell 2021; 34:1532-1544. [PMID: 34117611 DOI: 10.1007/s13577-021-00557-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
Cell death pathways related to ferroptosis are implicated in the progression of melanoma. Emerging data reporting the upregulation of microRNA (miR)-130b-3p in melanoma indicate the potential implication of miR-130b-3p in this malignancy. Herein, we aimed to identify whether and how miR-130b-3p regulated ferroptosis in melanoma cells. Melanoma cells (A375, G-361) were treated with erastin or RSL3 to mimic ferroptosis in vitro. Viability, lipid peroxidation level and ferrous ion content in melanoma cells were then assessed in response to manipulation of miR-130b-3p expression. Luciferase assay was conducted to determine the binding of miR-130b-3p to Dickkopf1 (DKK1). Western blot assay was conducted to determine the expression of molecules related to nuclear factor-erythroid 2 p45-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. The results indicated that miR-130b-3p exerted an inhibitory role in erastin or RSL3-induced ferroptosis, evidenced by reductions in lipid peroxidation and ferrous ion content. By suppressing the expression of target gene DKK1, miR-130b-3p activated the Nrf2/HO-1 pathway, whereby repressing ferroptosis. miR-130b-3p blocked the antitumor activity of erastin. Further, in vitro findings were reproduced in an in vivo murine model. Together, these data suggest the potential of miR-130b-3p to inhibit ferroptosis in melanoma cells and the mechanism was related to DKK1-mediated Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Yangying Liao
- Department of Dermatology, Hunan Provincial People's HospitalThe First Affiliated Hospital of Hunan Normal University)Hunan Province, No. 61, Jiefang West Road, Changsha, 410005, People's Republic of China
| | - Xiaomin Jia
- Department of Pathology, Lhasa People's Hospital of Tibet Autonomous Region, Lhasa, 850000, People's Republic of China
| | - Yi Ren
- Beijing Jishuitan Hospital, Beijing, 100035, People's Republic of China
| | - Zhuoga Deji
- Department of Pathology, Lhasa People's Hospital of Tibet Autonomous Region, Lhasa, 850000, People's Republic of China
| | - Yuzhen Gesang
- Department of Pathology, Lhasa People's Hospital of Tibet Autonomous Region, Lhasa, 850000, People's Republic of China
| | - Ning Ning
- Medical Department, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, People's Republic of China
| | - Hao Feng
- Department of Dermatology, Hunan Provincial People's HospitalThe First Affiliated Hospital of Hunan Normal University)Hunan Province, No. 61, Jiefang West Road, Changsha, 410005, People's Republic of China.
| | - Hong Yu
- Department of Pathology, The Third People's Hospital of Shenzhen, Shenzhen, 518100, People's Republic of China
| | - An Wei
- Department of Ultrasound, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61, Jiefang West Road, Changsha, 410005, Hunan Province, People's Republic of China.
| |
Collapse
|
39
|
Singh P, Chen Y, Tyagi D, Wu L, Ren X, Feng J, Carrier A, Luan T, Tang Y, Zhang J, Zhang X. β-Cyclodextrin-grafted hyaluronic acid as a supramolecular polysaccharide carrier for cell-targeted drug delivery. Int J Pharm 2021; 602:120602. [PMID: 33862128 DOI: 10.1016/j.ijpharm.2021.120602] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/03/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023]
Abstract
β-Cyclodextrin (β-CD) was grafted onto hyaluronic acid (HA) in a single step to generate a supramolecular biopolymer (HA-β-CD) that was explored for targeted drug delivery applications. Along with its excellent biocompatibility, the prepared HA-β-CD exhibits not only exceptionally high loading capacity for the model drugs doxorubicin and Rhodamine B through the formation of inclusion complexes with the β-CD component, but also the capability of targeted drug delivery to cancerous cells with a high level of expression of CD44 receptors, attributable to its HA component. The polymer can release the drug under slightly acidic conditions. With all its attributes, HA-β-CD may be a promising cancer-cell-targeting drug carrier.
Collapse
Affiliation(s)
- Parbeen Singh
- Postdoctoral Innovation Practice Base, Department of Biological Applied Engineering, Shenzhen Key Laboratory of Fermentation, Purification and Analysis, Shenzhen Polytechnic, Shenzhen 518055, China; State Key Laboratory Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou 51027, China; Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongli Chen
- Postdoctoral Innovation Practice Base, Department of Biological Applied Engineering, Shenzhen Key Laboratory of Fermentation, Purification and Analysis, Shenzhen Polytechnic, Shenzhen 518055, China; State Key Laboratory Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou 51027, China
| | - Deependra Tyagi
- School of Basic Medical Sciences, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Li Wu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaohong Ren
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jinglong Feng
- Postdoctoral Innovation Practice Base, Department of Biological Applied Engineering, Shenzhen Key Laboratory of Fermentation, Purification and Analysis, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Andrew Carrier
- Department of Chemistry and Department of Health Sciences, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| | - Tiangang Luan
- State Key Laboratory Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou 51027, China
| | - Yongjun Tang
- Postdoctoral Innovation Practice Base, Department of Biological Applied Engineering, Shenzhen Key Laboratory of Fermentation, Purification and Analysis, Shenzhen Polytechnic, Shenzhen 518055, China.
| | - Jiwen Zhang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, No.2 Tiantan Xili, Beijing 100050, China.
| | - Xu Zhang
- Department of Chemistry and Department of Health Sciences, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada.
| |
Collapse
|
40
|
Bernhard S, Tibbitt MW. Supramolecular engineering of hydrogels for drug delivery. Adv Drug Deliv Rev 2021; 171:240-256. [PMID: 33561451 DOI: 10.1016/j.addr.2021.02.002] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
Supramolecular binding motifs are increasingly employed in the design of biomaterials. The ability to rationally engineer specific yet reversible associations into polymer networks with supramolecular chemistry enables injectable or sprayable hydrogels that can be applied via minimally invasive administration. In this review, we highlight two main areas where supramolecular binding motifs are being used in the design of drug delivery systems: engineering network mechanics and tailoring drug-material affinity. Throughout, we highlight many of the established and emerging chemistries or binding motifs that are useful for the design of supramolecular hydrogels for drug delivery applications.
Collapse
|
41
|
Chen X, Wang M, Hu Y, Gong T, Zhang ZR, Yu R, Fu Y. Low-dose paclitaxel via hyaluronan-functionalized bovine serum albumin nanoparticulate assembly for metastatic melanoma treatment. J Mater Chem B 2021; 8:2139-2147. [PMID: 32090232 DOI: 10.1039/c9tb02780g] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Due to the critical role of CD44 in mediating cell adhesion and migration, CD44-targeted drug delivery via hyaluronan has been extensively explored. Herein, cationic bovine serum albumin nanoparticles were assembled with hyaluronan (HA) of various molecular weights via simple electrostatic interaction to afford hierarchical nanoparticles (HNPs) with various size distributions and structures. Next, HNPs obtained using 49 kDa HA have been used to encapsulate paclitaxel (PTX-HNPs), which demonstrated selective lung accumulation due to both size effect and CD44-mediated targetability. Biodistribution studies showed that HNPs enhanced the lung specific accumulation of HNPs in the C57BL/6 mice melanoma lung metastasis model. In the antitumor studies, compared with the Taxol or bovine serum albumin nanoparticle (NP) groups, PTX-HNPs significantly inhibited B16F10 lung metastasis at a relatively low dose. Additionally, cell migration and invasion experiments in vitro further confirmed that PTX-HNPs significantly inhibited the migration of B16F10 cells compared to Taxol or paclitaxel-loaded NP groups. Overall, our results suggest that PTX-HNPs represent a highly promising strategy for the treatment of lung metastatic melanoma.
Collapse
Affiliation(s)
- Xue Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Mou Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Ying Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Zhi-Rong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Ruilian Yu
- Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
42
|
Liu Y, Tiruthani K, Wang M, Zhou X, Qiu N, Xiong Y, Pecot CV, Liu R, Huang L. Tumor-targeted gene therapy with lipid nanoparticles inhibits tumor-associated adipocytes and remodels the immunosuppressive tumor microenvironment in triple-negative breast cancer. NANOSCALE HORIZONS 2021; 6:319-329. [PMID: 33587080 PMCID: PMC8638658 DOI: 10.1039/d0nh00588f] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Adipocytes are the primary cellular components within the tumor microenvironment (TME) of triple-negative breast cancer (TNBC). Increasing evidence suggests that tumor-associated adipocytes (TAAs) can aggravate tumor progression, exacerbate the immunosuppressive TME and compromise therapeutic efficacy. In this study, the biological effect of TAAs within the breast cancer TME is first investigated, and the C-C Motif Chemokine Ligand 2 (CCL2) which is mainly secreted by TAAs in the extracellular environment is identified as the key mediator. CCL2 recruits immune cells such as monocytes and macrophages that further differentiated into immunosuppressive myeloid-derived suppressor cells (MDSCs) and M2 macrophages. To manipulate CCL2-mediated immune response, a protein trap that binds with CCL2 with high affinity and specificity is designed. The plasmid DNA encoding the CCL2 trap (pCCL2) is specifically delivered to the TME by using targeted lipid-protamine-DNA (LPD) nanoparticles to locally express the CCL2 trap and ameliorate the immunosuppressive TME. Significantly, compared with the commercially available CCL2 antibody, this strategy shows enhanced therapeutic efficacy and appreciable tumor growth inhibition. Furthermore, the pCCL2 trap treatment successfully suppresses TAAs, increases T cell infiltration and decreases the population of immunosuppressive M2 macrophages and MDSCs. Further studies show that the pCCL2 trap could facilitate PD-L1 blockade immunotherapy, demonstrating its translation potential.
Collapse
Affiliation(s)
- Yun Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karthik Tiruthani
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Menglin Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xuefei Zhou
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nasha Qiu
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yang Xiong
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chad V. Pecot
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rihe Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
43
|
Dual Encapsulated Dacarbazine and Zinc Phthalocyanine Polymeric Nanoparticle for Photodynamic Therapy of Melanoma. Pharm Res 2021; 38:335-346. [PMID: 33604784 DOI: 10.1007/s11095-021-02999-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 11/05/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Melanoma is an invasive and very aggressive skin cancer due to its multi-drug resistance that results in poor patient survival. There is a need to test new treatment approaches to improve therapeutic efficacy and reduce side effects of conventional treatments. METHODS PLA/PVA nanoparticles carrying both Dacarbazine and zinc phthalocyanine was produced by double emulsion technique. The characterization was performed by dynamic light scattering and atomic force microscopy. In vitro photodynamic therapy test assay using MV3 melanoma cells as a model has been performed. In vitro cell viability (MTT) was performed to measure cell toxicity of of nanoparticles with and without drugs using human endothelial cells as a model. The in vivo assay (biodistribution/tissue deposition) has been performed using radiolabeled PLA/PVA NPs. RESULTS The nanoparticles produced showed a mean diameter of about 259 nm with a spherical shape. The in-vitro photodynamic therapy tests demonstrated that the combination is critical to enhance the therapeutic efficacy and it is dose dependent. The in vitro cell toxicity assay using endothelial cells demonstrated that the drug encapsulated into nanoparticles had no significant toxicity compared to control samples. In-vivo results demonstrated that the drug loading affects the biodistribution of the nanoparticle formulations (NPs). Low accumulation of the NPs into the stomach, heart, brain, and kidneys suggested that common side effects of Dacarbazine could be reduced. CONCLUSION This work reports a robust nanoparticle formulation with the objective to leveraging the synergistic effects of chemo and photodynamic therapies to potentially suppressing the drug resistance and reducing side effects associated with Dacarbazine. The data corroborates that the dual encapsulated NPs showed better in-vitro efficacy when compared with the both compounds alone. The results support the need to have a dual modality NP formulation for melanoma therapy by combining chemotherapy and photodynamic therapy.
Collapse
|
44
|
Wang Y, Shen N, Wang Y, Zhang Y, Tang Z, Chen X. Self-Amplifying Nanotherapeutic Drugs Homing to Tumors in a Manner of Chain Reaction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2002094. [PMID: 33382144 DOI: 10.1002/adma.202002094] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 11/26/2020] [Indexed: 06/12/2023]
Abstract
Active tumor-targeting drug delivery has great potency in cancer therapy. However, the targeting efficiency of traditional active tumor-targeting nanotherapeutic drugs is limited by the scarcity of their accessible targets/receptors in tumors. Here, a novel self-amplifying tumor-targeting strategy with a chain reaction mechanism is developed. A coagulation targeting peptide (GNQEQVSPLTLLKXC, termed A15)-decorated poly(L-glutamic acid)-graft-maleimide poly(ethylene glycol)/combretastatin A4 conjugate (A15-PLG-CA4) is prepared to obtain a self-amplifying nanotherapeutic platform homing to tumors. After administration to tumor-bearing mice, A15-PLG-CA4 starts a chain reaction cycle consisting of intratumoral hemorrhage, target FXIIIa amplification, blood clot binding, and CA4 release in tumors. In this way, A15-PLG-CA4 increases the level of its accessible targets (FXIIIa) in a manner of chain reaction. The FXIIIa activity at 8 h is 4.1-fold more than the one at 0 h in the C26 tumors treated with A15-PLG-CA4. The total CA4 concentration at 24 h is 2.9-fold more than the control. A15-PLG-CA4 shows a significantly higher antitumor effect against large C26 tumors (≈500 mm3 ) thanks to the remarkable tumor-targeting ability compared with the control. Therefore, this report highlights the potential of the self-amplifying tumor-targeting strategy in the development of next generation active tumor-targeting nanotherapeutic drugs for tumor therapy.
Collapse
Affiliation(s)
- Yue Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100039, P. R. China
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Ying Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
45
|
Wang Y, Ren S, Gong X, Wang J, Zhu N, Cai D, Ruan J. Prognostic factors for postoperative survival in melanoma patients with bone metastasis. Medicine (Baltimore) 2021; 100:e24558. [PMID: 33530285 PMCID: PMC7850704 DOI: 10.1097/md.0000000000024558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 01/12/2021] [Indexed: 01/05/2023] Open
Abstract
Melanoma can spread to the bone by metastasis and is relevant to a poor outcome. However, because of the rarity of melanoma patients with bone metastasis, the prognostic postoperative survival factors of them have not been elucidated. The aim of this special population-based cohort was to elucidate the prognostic factors associated with postoperative survival. The Surveillance, Epidemiology, and End Results database was used to extract postoperative survival data relating to patients with melanoma and bone metastasis at diagnosis between 2010 and 2016, along with data on a range of potential postoperative prognostic factors. We then investigated the potential postoperative prognostic roles of these factors using a Cox regression model and the Kaplan-Meier analysis. In all, the Surveillance, Epidemiology, and End Results database included 186 cases. Regarding overall survival, the 1-, 3-, and 5-year overall survival rates for the entire cohort were 36.2%, 15.4%, and 9.5%, respectively. Regarding cancer-specific survival, the 1-, 3-, and 5-year cancer-specific survival rates were 42.0%, 23.2%, and 16.6%, respectively. Within a cohort of melanoma patients with bone metastasis after surgery, our analysis showed that a smaller tumor size and the lack of metastases at other sites were predictors of survival.
Collapse
Affiliation(s)
- Yucheng Wang
- Department of Orthopedics, Taizhou University Affiliated Municipal Hospital, Taizhou, Zhejiang
- Hebei North University, Zhangjiakou, Hebei
| | - Shihong Ren
- Department of Orthopedics, The First People's Hospital of Wenling, Taizhou, Zhejiang, China
| | - Xiaokang Gong
- Department of Orthopedics, Taizhou University Affiliated Municipal Hospital, Taizhou, Zhejiang
| | - Jiacheng Wang
- Department of Orthopedics, Taizhou University Affiliated Municipal Hospital, Taizhou, Zhejiang
| | - Ning Zhu
- Department of Orthopedics, Taizhou University Affiliated Municipal Hospital, Taizhou, Zhejiang
| | | | - Jianwei Ruan
- Department of Orthopedics, Taizhou University Affiliated Municipal Hospital, Taizhou, Zhejiang
| |
Collapse
|
46
|
Mancuso A, Cristiano MC, Fresta M, Paolino D. The Challenge of Nanovesicles for Selective Topical Delivery for Acne Treatment: Enhancing Absorption Whilst Avoiding Toxicity. Int J Nanomedicine 2020; 15:9197-9210. [PMID: 33239876 PMCID: PMC7682599 DOI: 10.2147/ijn.s237508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/24/2020] [Indexed: 12/18/2022] Open
Abstract
Acne is a common skin disease that affect over 80% of adolescents. It is characterized by inflammation of the hair bulb and the attached sebaceous gland. To date, many strategies have been used to treat acne as a function of the disease severity. However, common treatments for acne seem to show several side effects, from local irritation to more serious collateral effects. The use of topical vesicular carriers able to deliver active compounds is currently considered as an excellent approach in the treatment of different skin diseases. Many results in the literature have proven that drug delivery systems are useful in overcoming the toxicity induced by common drug therapies, while maintaining their therapeutic efficacy. Starting from these assumptions, the authors reviewed drug delivery systems already realized for the topical treatment of acne, with a focus on their limitations and advantages over conventional treatment strategies. Although their exact mechanism of permeation is not often completely clear, deformable vesicles seem to be the best solution for obtaining a specific delivery of drugs into the deeper skin layers, with consequent increased local action and minimized collateral effects.
Collapse
Affiliation(s)
- Antonia Mancuso
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro88100, Italy
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, Catanzaro88100, Italy
| | - Massimo Fresta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro88100, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, Catanzaro88100, Italy
| |
Collapse
|
47
|
Cordeiro AP, Feuser PE, Figueiredo PG, Cunha ESD, Martinez GR, Machado-de-Ávila RA, Rocha MEM, Araújo PHHD, Sayer C. In vitro synergic activity of diethyldithiocarbamate and 4-nitrochalcone loaded in beeswax nanoparticles against melanoma (B16F10) cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 120:111651. [PMID: 33545819 DOI: 10.1016/j.msec.2020.111651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/03/2020] [Accepted: 10/13/2020] [Indexed: 11/27/2022]
Abstract
The use of nanoparticles as drug delivery systems to simultaneously carry several therapeutic agents is an attractive idea to create new synergic treatments and to develop the next generation of cancer therapies. Therefore, the goal of this study was the simultaneous encapsulation of a hydrophilic drug, sodium diethyldithiocarbamate (DETC), and a hydrophobic drug, 4-nitrochalcone (4NC), in beeswax nanoparticles (BNs) to evaluate the in vitro synergic activity of this combination against melanoma (B16F10) cells. BNs were prepared by water/oil/water double emulsion in the absence of organic solvents. Transmission electron microscopy imaging and dynamic light scattering analyses indicated the formation of BNs with a semispherical shape, average diameter below 250 nm, relatively narrow distributions, and negative zeta potential. The double emulsion technique proved to be effective for the simultaneous encapsulation of DETC and 4NC with efficiencies of 86.2% and 98.7%, respectively, and this encapsulation did not affect the physicochemical properties of the BNs. DETC and 4NC loaded in BNs exhibited a higher cytotoxicity toward B16F10 cells than free 4NC and DETC. This simultaneous encapsulation led to a synergic effect of DETC and 4NC on B16F10 cells, decreasing the cell viability from 46% (DETC BNs) and 54% (4NC BNs) to 64% (DETC+4NC BNs). Therefore, the IC50 of DETC+4NC was also lower than that of either when individually encapsulated, and that of free DETC or 4NC. Therefore, DETC and 4NC were efficiently simultaneously encapsulated in BNs and this drug combination was able to generate an in vitro synergic therapeutic effect on B16F10 cells.
Collapse
Affiliation(s)
- Arthur Poester Cordeiro
- Department of Chemical and Food Engineering, Federal University of Santa Catarina, SC, Brazil
| | - Paulo Emílio Feuser
- Department of Chemical and Food Engineering, Federal University of Santa Catarina, SC, Brazil
| | | | | | | | | | | | | | - Claudia Sayer
- Department of Chemical and Food Engineering, Federal University of Santa Catarina, SC, Brazil.
| |
Collapse
|
48
|
Gu TW, Wang MZ, Niu J, Chu Y, Guo KR, Peng LH. Outer membrane vesicles derived from E. coli as novel vehicles for transdermal and tumor targeting delivery. NANOSCALE 2020; 12:18965-18977. [PMID: 32914815 DOI: 10.1039/d0nr03698f] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Transdermal drug delivery is favored in clinical therapy because of its ability to overcome the shortcomings of the first pass elimination of the liver caused by traditional oral administration and the irreversibility of the injection. However, skin stratum corneum (SC) forms a big barrier that precludes most of the biomacromolecules. Herein, we propose the engineering of transformed Escherichia coli (E. coli) derived outer membrane vesicles, detoxified by lysozymes (named TEVs) as the carrier for transdermal drug delivery. TEVs were derived from transgenic E. coli and then modified by an integrin alpha(v)beta(3) (αvβ3) targeting peptide and co-loaded with indocyanine green (ICG) (P-TEVs-G). TEVs were shown to have excellence in penetrating through intact SC without any additional enhancement, followed by targeting of melanoma cells. TEVs are promising nanoplatforms for transdermal and tumor targeting drug delivery with high efficacy and biosafety, possessing great potential in the treatment of superficial tumors.
Collapse
Affiliation(s)
- Ting-Wei Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China.
| | | | | | | | | | | |
Collapse
|
49
|
Ferraz CAA, de Oliveira Júnior RG, de Oliveira AP, Groult H, Beaugeard L, Picot L, de Alencar Filho EB, Almeida JRGDS, Nunes XP. Complexation with β-cyclodextrin enhances apoptosis-mediated cytotoxic effect of harman in chemoresistant BRAF-mutated melanoma cells. Eur J Pharm Sci 2020; 150:105353. [PMID: 32334103 DOI: 10.1016/j.ejps.2020.105353] [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: 02/11/2020] [Revised: 03/27/2020] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
Harman, a natural β-carboline alkaloid, has recently gained considerable interest due to its anticancer properties. However, its physicochemical characteristics and poor oral bioavailability have been limiting factors for its pharmaceutical development. In this paper, we described the complexation of harman (HAR) with β-cyclodextrin (βCD) as a promising alternative to improve its solubility and consequently its cytotoxic effect in chemoresistant melanoma cells (A2058 cell line). Inclusion complexes (βCD-HAR) were prepared using a simple method and then characterized by FTIR, NMR and SEM techniques. Through in silico studies, the mechanism of complexation of HAR with βCD was elucidated in detail. Both HAR and βCD-HAR promoted cytotoxicity, apoptosis, cell cycle arrest and inhibition of cell migration in melanoma cells. Interestingly, complexation of HAR with βCD enhanced its pro-apoptotic effect by increasing of caspase-3 activity (p < 0.05), probably due to an improvement in HAR solubility. In addition, HAR and βCD-HAR sensitized A2058 cells to vemurafenib, dacarbazine and 5FU treatments, potentializing their cytotoxic activity. These findings suggest that complexation of HAR with natural polymers such as βCD can be useful to improve its bioavailability and antimelanoma activity.
Collapse
Affiliation(s)
- Christiane Adrielly Alves Ferraz
- NEPLAME, Universidade Federal do Vale do São Francisco, Petrolina-PE, 56306-000, Brazil; RENORBIO, Universidade Federal Rural de Pernambuco, Recife-PE, 52171-900, Brazil
| | | | - Ana Paula de Oliveira
- NEPLAME, Universidade Federal do Vale do São Francisco, Petrolina-PE, 56306-000, Brazil
| | - Hugo Groult
- UMRi CNRS 7266 LIENSs, La Rochelle Université, La Rochelle, 17042, France
| | - Laureen Beaugeard
- UMRi CNRS 7266 LIENSs, La Rochelle Université, La Rochelle, 17042, France
| | - Laurent Picot
- UMRi CNRS 7266 LIENSs, La Rochelle Université, La Rochelle, 17042, France
| | | | | | - Xirley Pereira Nunes
- NEPLAME, Universidade Federal do Vale do São Francisco, Petrolina-PE, 56306-000, Brazil.
| |
Collapse
|
50
|
Brachytherapy in a Single Dose of 10Gy as an "in situ" Vaccination. Int J Mol Sci 2020; 21:ijms21134585. [PMID: 32605154 PMCID: PMC7369911 DOI: 10.3390/ijms21134585] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) is one of the major methods of cancer treatment. RT destroys cancer cells, but also affects the tumor microenvironment (TME). The delicate balance between immunomodulation processes in TME is dependent, among other things, on a specific radiation dose. Despite many studies, the optimal dose has not been clearly determined. Here, we demonstrate that brachytherapy (contact radiotherapy) inhibits melanoma tumor growth in a dose-dependent manner. Doses of 10Gy and 15Gy cause the most effective tumor growth inhibition compared to the control group. Brachytherapy, at a single dose of ≥ 5Gy, resulted in reduced tumor blood vessel density. Only a dose of 10Gy had the greatest impact on changes in the levels of tumor-infiltrating immune cells. It most effectively reduced the accumulation of protumorogenic M2 tumor-associated macrophages and increased the infiltration of cytotoxic CD8+ T lymphocytes. To summarize, more knowledge about the effects of irradiation doses in anticancer therapy is needed. It may help in the optimization of RT treatment. Our results indicate that a single dose of 10Gy leads to the development of a robust immune response. It seems that it is able to convert a tumor microenvironment into an “in situ” vaccine and lead to a significant inhibition of tumor growth.
Collapse
|