1
|
Gulati S, Ansari N, Moriya Y, Joshi K, Prasad D, Sajwan G, Shukla S, Kumar S, Varma RS. Nanobiopolymers in cancer therapeutics: advancing targeted drug delivery through sustainable and controlled release mechanisms. J Mater Chem B 2024; 12:11887-11915. [PMID: 39502076 DOI: 10.1039/d4tb00599f] [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: 11/28/2024]
Abstract
Nanobiopolymers have emerged as a transformative frontier in cancer treatment, leveraging nanotechnology to transform drug delivery. This review provides a comprehensive exploration of the multifaceted landscape of nano-based biopolymers, emphasizing their diverse sources, synthesis methods, and classifications. Natural, synthetic, and microbial nanobiopolymers are scrutinized, along with elucidation of their underlying mechanisms and impact on cancer drug delivery; the latest findings on their deployment as targeted drug delivery agents for cancer treatment are discussed. A detailed analysis of nanobiopolymer sources, including polysaccharides, peptides, and nucleic acids, highlights critical attributes like biodegradability, renewability, and sustainability essential for therapeutic applications. The classification of nanobiopolymers based on their origin and differentiation among natural, synthetic, and microbial sources are thoroughly examined for inherent advantages, challenges, and suitability for cancer therapeutics. The importance of targeted drug release at tumour sites, crucial for minimizing adverse effects on normal tissues, is discussed, encompassing various mechanisms. The role of polymer membrane coatings as a pivotal barrier for facilitating controlled drug release through diffusion is elucidated, providing further insight into efficient methods for cancer treatment and thus consolidating the current knowledge base for researchers and practitioners in the field of nanobiopolymers and cancer therapeutics.
Collapse
Affiliation(s)
- Shikha Gulati
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India.
| | - Nabeela Ansari
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India
| | - Yamini Moriya
- Department of Life Sciences, Sri Venkateswara College, University of Delhi, Delhi-110021, India
| | - Kumud Joshi
- Department of Life Sciences, Sri Venkateswara College, University of Delhi, Delhi-110021, India
| | - Disha Prasad
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India.
| | - Gargi Sajwan
- Department of Biological Sciences, Sri Venkateswara College, University of Delhi, Delhi-110021, India
| | - Shefali Shukla
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India.
| | - Sanjay Kumar
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi-110021, India.
| | - Rajender S Varma
- Centre of Excellence for Research in Sustainable Chemistry, Department of Chemistry, Federal University of São Carlos, 13565-905 São Carlos - SP, Brazil.
| |
Collapse
|
2
|
Rathore SS, Leno Jenita JJ, Dotherabandi M. A systematic review on hyaluronic acid coated nanoparticles: recent strategy in breast cancer management. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-42. [PMID: 39429014 DOI: 10.1080/09205063.2024.2416293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Hyaluronic acid, a non-sulphated glycosaminoglycan has attracted its usage in the management of breast cancer. Drug-loaded nanoparticles with hyaluronic acid surface modifications show potential as a promising method for targeting and delivering drugs to the tumor site. The aim of this study was to conduct a systematic review of articles and assess the impact of hyaluronic acid coated nanoparticles on breast cancer. The various database were used for this comprehensive review. The inclusion and exclusion criteria were selected according to the PRISMA guidelines. Studies associated with characterization, in vitro, and in vivo studies were collected and subjected for further analysis. According to the inclusion criteria, 41 literature were selected for analysis. From all the studies, it was observed that the nanoparticles coated with hyaluronic acid produced better particle size, shape, zeta potential, increased in vitro cytotoxicity, cellular uptake, cell apoptosis, and anti-tumor effect in vivo. Research has shown that hyaluronic acid exhibits a higher affinity for CD44 receptors, resulting in enhanced targeted nanoparticle activity on cancer cells while sparing normal cells.
Collapse
Affiliation(s)
- Seema S Rathore
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bangalore, India
| | - J Josephine Leno Jenita
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bangalore, India
| | - Manjula Dotherabandi
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bangalore, India
| |
Collapse
|
3
|
Chen A, Zhu W, Goding R. Analysis of naproxen activation of cell death pathways in Colo320 cells. Mol Clin Oncol 2024; 21:61. [PMID: 39071975 PMCID: PMC11273252 DOI: 10.3892/mco.2024.2759] [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: 09/17/2023] [Accepted: 06/07/2024] [Indexed: 07/30/2024] Open
Abstract
Colorectal cancer is a life-threatening and prevalent type of cancer. However, a number of current treatments have serious side effects, which increase the need for alternatives. Non-steroidal anti-inflammatory drugs have potential chemopreventive capabilities. The present study aimed to confirm this, as well as to investigate potential pathways and reasons for this trait. To accomplish this, cancerous Colo320 and healthy CCD-18 cells were treated with various concentrations of naproxen sodium (NS). A caspase-3 assay revealed a statistically significant increase in caspase-3 activity in Colo320 cells (300%; P<0.01), but not in CCD-18 cells. This chemical was also associated with a significant decrease in Colo320 cell survival (-72.888%; P<0.01), but not CCD-18 cell survival. Furthermore, NS was found to significantly decrease the migration of Colo320 cells (86.58%; P<0.01). Finally, RNA sequencing of cells treated with NS revealed the statistically significant downregulation of the mucin 5B, oligomeric mucus/gel-forming, S100 calcium binding protein A9 and mucin 5AC, oligomeric mucus/gel-forming genes, which are upregulated in colorectal cancer and are known to contribute to cancer proliferation, stemness and drug resistance. These novel biological pathway results were further confirmed using ELISAs. The present study identified a novel molecular mechanism of the anti-colorectal cancer activity of NS.
Collapse
Affiliation(s)
- Andrew Chen
- College of Arts and Sciences at The State University of New York at Stony Brook, Stony Brook, NY 11794, USA
- SCI Research Institute, Jericho, NY 11753, USA
| | - Wei Zhu
- SCI Research Institute, Jericho, NY 11753, USA
- Neuroscience Research Institute at The State University of New York at Old Westbury, Old Westbury, NY 11568, USA
| | - Rian Goding
- SCI Research Institute, Jericho, NY 11753, USA
- College of Osteopathic Medicine at The New York Institute of Technology, Old Westbury, NY 11568, USA
| |
Collapse
|
4
|
Kareemi AF, Likhitkar S. Applications and advancements of polysaccharide-based nanostructures for enhanced drug delivery. Colloids Surf B Biointerfaces 2024; 238:113883. [PMID: 38615389 DOI: 10.1016/j.colsurfb.2024.113883] [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: 02/01/2024] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
Growing demand for highly effective, site-specific delivery of pharmaceuticals and nutraceuticals using nano-sized carriers has prompted increased scrutiny of carrier biocompatibility and biodegradability. To address these concerns, biodegradable natural polymers have emerged as a transformative domain, offering non-toxic, precisely targetable carriers capable of finely modulating cargo pharmacokinetics while generating innocuous decomposition by-products. This comprehensive review illuminates the emergence of polysaccharide-based nanoparticulate drug delivery systems. These systems establish an interactive interface between drug and targeted organs, guided by strategic modifications to polysaccharide backbones, which facilitate the creation of morphologically, constitutionally, and characteristically vibrant nanostructures through various fabrication routes, underpinning their pivotal role in biomedical applications. Advancements crucial to enhancing polysaccharide-based drug delivery, such as surface modifications and bioinspired modifications for enhanced targeting, and stimuli-responsive release, strategies to overcome biological barriers, enhance tumor penetration, and optimize therapeutic outcomes are highlighted. This review also examines some potent challenges, and the contemporary way out of them, and discusses future perspectives in the field.
Collapse
Affiliation(s)
- Asra Fatimah Kareemi
- Department of Chemistry, St. Aloysius College (Autonomous), Jabalpur, Madhya Pradesh 482001, India
| | - Sweta Likhitkar
- Department of Chemistry, St. Aloysius College (Autonomous), Jabalpur, Madhya Pradesh 482001, India.
| |
Collapse
|
5
|
Valverde TM, dos Santos VMR, Viana PIM, Costa GMJ, de Goes AM, Sousa LRD, Xavier VF, Vieira PMDA, de Lima Silva D, Domingues RZ, Ferreira JMDF, Andrade ÂL. Novel Fe 3O 4 Nanoparticles with Bioactive Glass-Naproxen Coating: Synthesis, Characterization, and In Vitro Evaluation of Bioactivity. Int J Mol Sci 2024; 25:4270. [PMID: 38673856 PMCID: PMC11049812 DOI: 10.3390/ijms25084270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Immune response to biomaterials, which is intimately related to their surface properties, can produce chronic inflammation and fibrosis, leading to implant failure. This study investigated the development of magnetic nanoparticles coated with silica and incorporating the anti-inflammatory drug naproxen, aimed at multifunctional biomedical applications. The synthesized nanoparticles were characterized using various techniques that confirmed the presence of magnetite and the formation of a silica-rich bioactive glass (BG) layer. In vitro studies demonstrated that the nanoparticles exhibited bioactive properties, forming an apatite surface layer when immersed in simulated body fluid, and biocompatibility with bone cells, with good viability and alkaline phosphatase activity. Naproxen, either free or encapsulated, reduced nitric oxide production, an inflammatory marker, while the BG coating alone did not show anti-inflammatory effects in this study. Overall, the magnetic nanoparticles coated with BG and naproxen showed promise for biomedical applications, especially anti-inflammatory activity in macrophages and in the bone field, due to their biocompatibility, bioactivity, and osteogenic potential.
Collapse
Affiliation(s)
- Thalita Marcolan Valverde
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (T.M.V.); (P.I.M.V.); (G.M.J.C.)
| | - Viviane Martins Rebello dos Santos
- Departamento de Química, Instituto de Ciências Exatas e Biológicas (ICEB), Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (V.M.R.d.S.); (D.d.L.S.)
| | - Pedro Igor Macário Viana
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (T.M.V.); (P.I.M.V.); (G.M.J.C.)
| | - Guilherme Mattos Jardim Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (T.M.V.); (P.I.M.V.); (G.M.J.C.)
| | - Alfredo Miranda de Goes
- Departamento de Patologia Geral, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| | - Lucas Resende Dutra Sousa
- Laboratório de Fitotecnologia, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (L.R.D.S.); (V.F.X.)
| | - Viviane Flores Xavier
- Laboratório de Fitotecnologia, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (L.R.D.S.); (V.F.X.)
| | - Paula Melo de Abreu Vieira
- Laboratório de Morfopatologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil;
| | - Daniel de Lima Silva
- Departamento de Química, Instituto de Ciências Exatas e Biológicas (ICEB), Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (V.M.R.d.S.); (D.d.L.S.)
| | - Rosana Zacarias Domingues
- Departamento de Química, Instituto de Ciências Exatas (ICEx), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| | - José Maria da Fonte Ferreira
- Departamento de Engenharia de Materiais e Cerâmica, CICECO, Universidade de Aveiro (UA), 3810193 Aveiro, Portugal;
| | - Ângela Leão Andrade
- Departamento de Química, Instituto de Ciências Exatas e Biológicas (ICEB), Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (V.M.R.d.S.); (D.d.L.S.)
- Departamento de Engenharia de Materiais e Cerâmica, CICECO, Universidade de Aveiro (UA), 3810193 Aveiro, Portugal;
| |
Collapse
|
6
|
Wang WD, Guo YY, Yang ZL, Su GL, Sun ZJ. Sniping Cancer Stem Cells with Nanomaterials. ACS NANO 2023; 17:23262-23298. [PMID: 38010076 DOI: 10.1021/acsnano.3c07828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cancer stem cells (CSCs) drive tumor initiation, progression, and therapeutic resistance due to their self-renewal and differentiation capabilities. Despite encouraging progress in cancer treatment, conventional approaches often fail to eliminate CSCs, necessitating the development of precise targeted strategies. Recent advances in materials science and nanotechnology have enabled promising CSC-targeted approaches, harnessing the power of tailoring nanomaterials in diverse therapeutic applications. This review provides an update on the current landscape of nanobased precision targeting approaches against CSCs. We elucidate the nuanced application of organic, inorganic, and bioinspired nanomaterials across a spectrum of therapeutic paradigms, encompassing targeted therapy, immunotherapy, and multimodal synergistic therapies. By examining the accomplishments and challenges in this potential field, we aim to inform future efforts to advance nanomaterial-based therapies toward more effective "sniping" of CSCs and tumor clearance.
Collapse
Affiliation(s)
- Wen-Da Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Yan-Yu Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Zhong-Lu Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Guang-Liang Su
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
7
|
Maghsoudi H, Sheikhnia F, Sitarek P, Hajmalek N, Hassani S, Rashidi V, Khodagholi S, Mir SM, Malekinejad F, Kheradmand F, Ghorbanpour M, Ghasemzadeh N, Kowalczyk T. The Potential Preventive and Therapeutic Roles of NSAIDs in Prostate Cancer. Cancers (Basel) 2023; 15:5435. [PMID: 38001694 PMCID: PMC10670652 DOI: 10.3390/cancers15225435] [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: 10/20/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Prostate cancer (PC) is the second most common type of cancer and the leading cause of death among men worldwide. Preventing the progression of cancer after treatments such as radical prostatectomy, radiation therapy, and hormone therapy is a major concern faced by prostate cancer patients. Inflammation, which can be caused by various factors such as infections, the microbiome, obesity and a high-fat diet, is considered to be the main cause of PC. Inflammatory cells are believed to play a crucial role in tumor progression. Therefore, nonsteroidal anti-inflammatory drugs along with their effects on the treatment of inflammation-related diseases, can prevent cancer and its progression by suppressing various inflammatory pathways. Recent evidence shows that nonsteroidal anti-inflammatory drugs are effective in the prevention and treatment of prostate cancer. In this review, we discuss the different pathways through which these drugs exert their potential preventive and therapeutic effects on prostate cancer.
Collapse
Affiliation(s)
- Hossein Maghsoudi
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Farhad Sheikhnia
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Przemysław Sitarek
- Department of Medical Biology, Medical University of Lodz, 90-151 Lodz, Poland
| | - Nooshin Hajmalek
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol 47176-47754, Iran;
| | - Sepideh Hassani
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Vahid Rashidi
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
| | - Sadaf Khodagholi
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada;
| | - Seyed Mostafa Mir
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49189-36316, Iran;
| | - Faezeh Malekinejad
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Fatemeh Kheradmand
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57147-83734, Iran
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57147-83734, Iran
| | - Mansour Ghorbanpour
- Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-88349, Iran;
| | - Navid Ghasemzadeh
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
| |
Collapse
|
8
|
Liu J, Chao T, Liu Y, Gong C, Zhang Y, Xiong H. Heterocyclic Molecular Targeted Drugs and Nanomedicines for Cancer: Recent Advances and Challenges. Pharmaceutics 2023; 15:1706. [PMID: 37376154 DOI: 10.3390/pharmaceutics15061706] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/28/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer is a top global public health concern. At present, molecular targeted therapy has emerged as one of the main therapies for cancer, with high efficacy and safety. The medical world continues to struggle with the development of efficient, extremely selective, and low-toxicity anticancer medications. Heterocyclic scaffolds based on the molecular structure of tumor therapeutic targets are widely used in anticancer drug design. In addition, a revolution in medicine has been brought on by the quick advancement of nanotechnology. Many nanomedicines have taken targeted cancer therapy to a new level. In this review, we highlight heterocyclic molecular-targeted drugs as well as heterocyclic-associated nanomedicines in cancer.
Collapse
Affiliation(s)
- Junxia Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yingying Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200000, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai 200000, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
9
|
Petrikaite V, D'Avanzo N, Celia C, Fresta M. Nanocarriers overcoming biological barriers induced by multidrug resistance of chemotherapeutics in 2D and 3D cancer models. Drug Resist Updat 2023; 68:100956. [PMID: 36958083 DOI: 10.1016/j.drup.2023.100956] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
Multidrug resistance (MDR) is currently a big challenge in cancer therapy and limits its success in several patients. Tumors use the MDR mechanisms to colonize the host and reduce the efficacy of chemotherapeutics that are injected as single agents or combinations. MDR mechanisms are responsible for inactivation of drugs and formbiological barriers in cancer like the drug efflux pumps, aberrant extracellular matrix, hypoxic areas, altered cell death mechanisms, etc. Nanocarriers have some potential to overcome these barriers and improve the efficacy of chemotherapeutics. In fact, they are versatile and can deliver natural and synthetic biomolecules, as well as RNAi/DNAi, thus providing a controlled release of drugs and a synergistic effect in tumor tissues. Biocompatible and safe multifunctional biopolymers, with or without specific targeting molecules, modify the surface and interface properties of nanocarriers. These modifications affect the interaction of nanocarriers with cellular models as well as the selection of suitable models for in vitro experiments. MDR cancer cells, and particularly their 2D and 3D models, in combination with anatomical and physiological structures of tumor tissues, can boost the design and preparation of nanomedicines for anticancer therapy. 2D and 3D cancer cell cultures are suitable models to study the interaction, internalization, and efficacy of nanocarriers, the mechanisms of MDR in cancer cells and tissues, and they are used to tailor a personalized medicine and improve the efficacy of anticancer treatment in patients. The description of molecular mechanisms and physio-pathological pathways of these models further allow the design of nanomedicine that can efficiently overcome biological barriers involved in MDR and test the activity of nanocarriers in 2D and 3D models of MDR cancer cells.
Collapse
Affiliation(s)
- Vilma Petrikaite
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių pr. 13, LT-50162 Kaunas, Lithuania; Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania.
| | - Nicola D'Avanzo
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy; Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy
| | - Christian Celia
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių pr. 13, LT-50162 Kaunas, Lithuania; Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy
| | - Massimo Fresta
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Viale "S. Venuta" s.n.c., 88100 Catanzaro, Italy
| |
Collapse
|
10
|
Huerta-Madroñal M, Espinosa-Cano E, Aguilar MR, Vazquez-Lasa B. Antiaging properties of antioxidant photoprotective polymeric nanoparticles loaded with coenzyme-Q10. BIOMATERIALS ADVANCES 2023; 145:213247. [PMID: 36527961 DOI: 10.1016/j.bioadv.2022.213247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 11/21/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Skin is the most extensive organ within our body. It is continually subjected to stress factors, among which ultraviolet irradiation, a key factor responsible in skin aging since it leads to reactive oxygen species production. In order to fight against these oxidative species, the human body has an innate robust antioxidant mechanism composed of several different substances, one of which is coenzyme Q10. Its capacity to increase cellular energy production and excellent antioxidant properties have been proved, as well as its antiaging properties being able to attenuate cellular damage induced by ultraviolet irradiation in human dermal fibroblasts. However, its high hydrophobicity and photolability hampers its therapeutic potential. In this context, the objective of this work consists of the preparation of chitosan-rosmarinic acid conjugate-based nanoparticles to encapsulate coenzyme Q10 with high encapsulation efficiencies in order to improve its bioavailability and broaden its therapeutic use in skin applications. Hyaluronic acid coating was performed giving stable nanoparticles at physiological pH with 382 ± 3 nm of hydrodynamic diameter (0.04 ± 0.02 polydispersity) and - 18 ± 3 mV of surface charge. Release kinetics studies showed a maximum of 82 % mass release of coenzyme Q10 after 40 min, and radical scavenger activity assay confirmed the antioxidant character of chitosan-rosmarinic acid nanoparticles. Hyaluronic acid-coated chitosan-rosmarinic acid nanoparticles loaded with coenzyme Q10 were biocompatible in human dermal fibroblasts and exhibited interesting photoprotective properties in ultraviolet irradiated cells. In addition, nanoparticles hindered the production of reactive oxygen species, interleukin-6 and metalloproteinase-1, as well as caspase-9 activation maintaining high viability values upon irradiation of dermal fibroblasts. Overall results envision a great potential of these nanovehicles for application in skin disorders or antiaging treatments.
Collapse
Affiliation(s)
- Miguel Huerta-Madroñal
- Group of Biomaterials, Institute of Polymer Science and Technology ICTP-CSIC, Madrid, Spain.
| | - Eva Espinosa-Cano
- Group of Biomaterials, Institute of Polymer Science and Technology ICTP-CSIC, Madrid, Spain.
| | - Maria Rosa Aguilar
- Group of Biomaterials, Institute of Polymer Science and Technology ICTP-CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain.
| | - Blanca Vazquez-Lasa
- Group of Biomaterials, Institute of Polymer Science and Technology ICTP-CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
11
|
Thermoresponsive in-situ gel containing hyaluronic acid and indomethacin for the treatment of corneal chemical burn. Int J Pharm 2023; 631:122468. [PMID: 36503038 DOI: 10.1016/j.ijpharm.2022.122468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/20/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
Ocular chemical burns are prevalent injuries that must have immediate and effective treatment to avoid complications. Aiming to improve bioavailability and efficacy, a poloxamer-based thermoresponsive in-situ gelling system containing hyaluronic acid and indomethacin was developed. Formulations with different polymeric proportions were screened through rheological measurements resulting in an optimized system (F2) with gelling temperature of 34.2 ± 0.11 °C. Its maximum viscosity varied from 77.33 mPa (25 °C) to 82.95 mPa (34 °C) following a non-Newtonian profile and a pH of 6.86 ± 0.01. No incompatibilities were found after infrared analysis. Polarized light microscopy and cryo-transmission electron microscopy have demonstrated micelles of nano-sized dimensions (21.86 nm) with indomethacin entrapped in the core, forming a polymeric network under heating. In vitro tests revealed a cumulative release of 59.75 ± 3.17 % up to 24 h under a sustained release profile. Results from HET-CAM assay indicated that F2 was well tolerated. Corneal wound healing was significantly faster in animals treated with F2 compared to a commercial formulation and an untreated group. These findings suggests that F2 could be an efficient system to delivery drugs into the ocular surface improving wound healing.
Collapse
|
12
|
Li C, Yu S, Chen J, Hou Q, Wang S, Qian C, Yin S. Risk stratification based on DNA damage-repair-related signature reflects the microenvironmental feature, metabolic status and therapeutic response of breast cancer. Front Immunol 2023; 14:1127982. [PMID: 37033959 PMCID: PMC10080010 DOI: 10.3389/fimmu.2023.1127982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
DNA damage-repair machinery participates in maintaining genomic integrity and affects tumorigenesis. Molecular signatures based on DNA damage-repair-related genes (DRGs) capable of comprehensively indicating the prognosis, tumor immunometabolic profile and therapeutic responsiveness of breast cancer (BRCA) patients are still lacking. Integrating public datasets and bioinformatics algorithms, we developed a robust prognostic signature based on 27 DRGs. Multiple patient cohorts identified significant differences in various types of survival between high- and low-risk patients stratified by the signature. The signature correlated well with clinicopathological factors and could serve as an independent prognostic indicator for BRCA patients. Furthermore, low-risk tumors were characterized by more infiltrated CD8+ T cells, follicular helper T cells, M1 macrophages, activated NK cells and resting dendritic cells, and fewer M0 and M2 macrophages. The favorable immune infiltration patterns of low-risk tumors were also accompanied by specific metabolic profiles, decreased DNA replication, and enhanced antitumor immunity. Low-risk patients may respond better to immunotherapy, and experience improved outcomes with conventional chemotherapy or targeted medicine. Real-world immunotherapy and chemotherapy cohorts verified the predictive results. Additionally, four small molecule compounds promising to target high-risk tumors were predicted. In vitro experiments confirmed the high expression of GNPNAT1 and MORF4L2 in BRCA tissues and their association with immune cells, and the knockdown of these two DRGs suppressed the proliferation of human BRCA cells. In summary, this DNA damage-repair-related signature performed well in predicting patient prognosis, immunometabolic profiles and therapeutic sensitivity, hopefully contributing to precision medicine and new target discovery of BRCA.
Collapse
Affiliation(s)
| | | | | | | | | | - Cheng Qian
- *Correspondence: Cheng Qian, ; Shulei Yin,
| | - Shulei Yin
- *Correspondence: Cheng Qian, ; Shulei Yin,
| |
Collapse
|
13
|
Nayak A, Warrier NM, Kumar P. Cancer Stem Cells and the Tumor Microenvironment: Targeting the Critical Crosstalk through Nanocarrier Systems. Stem Cell Rev Rep 2022; 18:2209-2233. [PMID: 35876959 PMCID: PMC9489588 DOI: 10.1007/s12015-022-10426-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2022] [Indexed: 11/25/2022]
Abstract
The physiological state of the tumor microenvironment (TME) plays a central role in cancer development due to multiple universal features that transcend heterogeneity and niche specifications, like promoting cancer progression and metastasis. As a result of their preponderant involvement in tumor growth and maintenance through several microsystemic alterations, including hypoxia, oxidative stress, and acidosis, TMEs make for ideal targets in both diagnostic and therapeutic ventures. Correspondingly, methodologies to target TMEs have been investigated this past decade as stratagems of significant potential in the genre of focused cancer treatment. Within targeted oncotherapy, nanomedical derivates-nanocarriers (NCs) especially-have emerged to present notable prospects in enhancing targeting specificity. Yet, one major issue in the application of NCs in microenvironmental directed therapy is that TMEs are too broad a spectrum of targeting possibilities for these carriers to be effectively employed. However, cancer stem cells (CSCs) might portend a solution to the above conundrum: aside from being quite heavily invested in tumorigenesis and therapeutic resistance, CSCs also show self-renewal and fluid clonogenic properties that often define specific TME niches. Further scrutiny of the relationship between CSCs and TMEs also points towards mechanisms that underly tumoral characteristics of metastasis, malignancy, and even resistance. This review summarizes recent advances in NC-enabled targeting of CSCs for more holistic strikes against TMEs and discusses both the current challenges that hinder the clinical application of these strategies as well as the avenues that can further CSC-targeting initiatives. Central role of CSCs in regulation of cellular components within the TME.
Collapse
Affiliation(s)
- Aadya Nayak
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Neerada Meenakshi Warrier
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Praveen Kumar
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
14
|
Jia Y, Chen S, Wang C, Sun T, Yang L. Hyaluronic acid-based nano drug delivery systems for breast cancer treatment: Recent advances. Front Bioeng Biotechnol 2022; 10:990145. [PMID: 36091467 PMCID: PMC9449492 DOI: 10.3389/fbioe.2022.990145] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer (BC) is the most common malignancy among females worldwide, and high resistance to drugs and metastasis rates are the leading causes of death in BC patients. Releasing anti-cancer drugs precisely to the tumor site can improve the efficacy and reduce the side effects on the body. Natural polymers are attracting extensive interest as drug carriers in treating breast cancer. Hyaluronic acid (HA) is a natural polysaccharide with excellent biocompatibility, biodegradability, and non-immunogenicity and is a significant component of the extracellular matrix. The CD44 receptor of HA is overexpressed in breast cancer cells and can be targeted to breast tumors. Therefore, many researchers have developed nano drug delivery systems (NDDS) based on the CD44 receptor tumor-targeting properties of HA. This review examines the application of HA in NDDSs for breast cancer in recent years. Based on the structural composition of NDDSs, they are divided into HA NDDSs, Modified HA NDDSs, and HA hybrid NDDSs.
Collapse
Affiliation(s)
- Yufeng Jia
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China
| | - Siwen Chen
- Center for Molecular Science and Engineering, College of Science, Northeastern University, Shenyang, China
- NHC Key Laboratory of Reproductive Health and Medical Genetics (China Medical University), Liaoning Research Institute of Family Planning (The Reproductive Hospital of China Medical University), Shenyang, China
| | - Chenyu Wang
- Department of Information Management, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Tao Sun
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China
- *Correspondence: Tao Sun, ; Liqun Yang,
| | - Liqun Yang
- NHC Key Laboratory of Reproductive Health and Medical Genetics (China Medical University), Liaoning Research Institute of Family Planning (The Reproductive Hospital of China Medical University), Shenyang, China
- *Correspondence: Tao Sun, ; Liqun Yang,
| |
Collapse
|
15
|
Abdelhaleem EF, Kassab AE, El-Nassan HB, Khalil OM. Design, synthesis, and biological evaluation of new celecoxib analogs as apoptosis inducers and cyclooxygenase-2 inhibitors. Arch Pharm (Weinheim) 2022; 355:e2200190. [PMID: 35976138 DOI: 10.1002/ardp.202200190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 11/06/2022]
Abstract
Series of new celecoxib analogs were synthesized to assess their anticancer activity against the MCF-7 cell line. Four compounds, 3a, 3c, 5b, and 5c, showed 1.4-9.2-fold more potent anticancer activity than celecoxib. The antiproliferative activity of the most potent compounds, 3c, 5b, and 5c, seems to be associated well with their ability to induce apoptosis in MCF-7 cells (18-24-fold). This evidence was supported by an increase in the expression of the tumor suppressor gene p53 (4-6-fold), the elevation in the Bax/BCL-2 ratio, and a significant increase in the level of active caspase-7 (4-7-fold). Moreover, compounds 3c and 5c showed significant cyclooxygenase-2 (COX-2) inhibitory activity. They were also docked into the crystal structure of the COX-2 enzyme (PDB ID: 3LN1) to understand their mode of binding.
Collapse
Affiliation(s)
- Eman F Abdelhaleem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala B El-Nassan
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Omneya M Khalil
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
16
|
Vetter VC, Wagner E. Targeting nucleic acid-based therapeutics to tumors: Challenges and strategies for polyplexes. J Control Release 2022; 346:110-135. [PMID: 35436520 DOI: 10.1016/j.jconrel.2022.04.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 12/18/2022]
Abstract
The current medical reality of cancer gene therapy is reflected by more than ten approved products on the global market, including oncolytic and other viral vectors and CAR T-cells as ex vivo gene-modified cell therapeutics. The development of synthetic antitumoral nucleic acid therapeutics has been proceeding at a lower but steady pace, fueled by a plethora of alternative nucleic acid platforms (from various antisense oligonucleotides, siRNA, microRNA, lncRNA, sgRNA, to larger mRNA and DNA) and several classes of physical and chemical delivery technologies. This review summarizes the challenges and strategies for tumor-targeted nucleic acid delivery. Focusing primarily on polyplexes (polycation complexes) as nanocarriers, delivery options across multiple barriers into tumor cells are illustrated.
Collapse
Affiliation(s)
- Victoria C Vetter
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany; Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany.
| |
Collapse
|
17
|
Abdelhaleem EF, Kassab AE, El-Nassan HB, Khalil OM. Design and Synthesis of Novel Celecoxib Analogues with Potential Cytotoxic and Pro-apoptotic Activity Against Breast Cancer Cell Line MCF-7. Med Chem 2022; 18:903-914. [PMID: 35264093 DOI: 10.2174/1573406418666220309123648] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/11/2022] [Accepted: 01/27/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Breast cancer is currently the leading cause of worldwide cancer incidence exceeding lung cancer. In addition, breast cancer accounts for 1 in 4 cancer cases and 1 in 6 cancer deaths among women. Cytotoxic chemotherapy is still the main therapeutic approach for patients with metastatic breast cancer. OBJECTIVE To synthesize a series of novel celecoxib analogues to evaluate their anticancer activity against MCF-7 cell line. METHOD Our design of target compounds was based on preserving the pyrazole moiety of celecoxib attached to two phenyl rings, one of them having polar hydrogen bonding group (sulfonamide or methoxy group). The methyl group of the second phenyl ring was replaced with chlorine or bromine atom. Finally, the trifluoromethyl group was replaced with arylidene hydrazine-1-carbonyl moiety, which is substituted either with fluoro or methoxy group, offering various electronic and lipophilic environments. These modifications were carried out to investigate their effects on the anti-proliferative activity of the newly synthesized celecoxib analogues and to provide a valuable structure activity relationship. RESULTS Four compounds namely (4e-h) exhibited significant antitumor activity. Compounds 4e, 4f and 4h showed 1.2-2 folds more potent anticancer activity than celecoxib. Celecoxib analogue 4f showed the most potent anti-proliferative activity. Its anti-proliferative activity seems to associate well with its ability to inhibit BCL-2. Moreover, activation of damage response pathway of the DNA leads to cell cycle arrest at G2/M phase, accumulation of cells in pre-G1 phase, indicating that cell death proceeds through an apoptotic mechanism. Compound 4f exhibited potent pro-apoptotic effect via induction of the intrinsic mitochondrial pathway of apoptosis. This mechanistic pathway was proved by a significant increase in the expression of the tumor suppressor gene p53, elevation in Bax/BCL-2 ratio and a significant increase in the level of active caspase-7. Furthermore, compound 4f showed moderate COX-2 inhibitory activity. CONCLUSION Celecoxib analogue 4f is a promising multi-targeted lead for the design and synthesis of potent anticancer agents.
Collapse
Affiliation(s)
- Eman F Abdelhaleem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Hala B El-Nassan
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Omneya M Khalil
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| |
Collapse
|
18
|
Kokoretsis D, Maniaki EK, Kyriakopoulou K, Koutsakis C, Piperigkou Z, Karamanos NK. Hyaluronan as "Agent Smith" in cancer extracellular matrix pathobiology: Regulatory roles in immune response, cancer progression and targeting. IUBMB Life 2022; 74:943-954. [PMID: 35261139 DOI: 10.1002/iub.2608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 12/16/2022]
Abstract
Extracellular matrix (ECM) critically regulates cancer cell behavior by governing cell signaling and properties. Hyaluronan (HA) acts as a structural and functional ECM component that mediates critical properties of cancer cells in a molecular size-dependent manner. HA fragments secreted by cancer-associated fibroblasts (CAFs) reveal the correlation of HA to CAF-mediated matrix remodeling, a key step for the initiation of metastasis. The main goal of this article is to highlight the vital functions of HA in cancer cell initiation and progression as well as HA-mediated paracrine interactions among cancer and stromal cells. Furthermore, the HA implication in mediating immune responses to cancer progression is also discussed. Novel data on the role of HA in the formation of pre-metastatic niche may contribute towards the improvement of current theranostic approaches that benefit cancer management.
Collapse
Affiliation(s)
- Dimitris Kokoretsis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Evangelia-Konstantina Maniaki
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
19
|
Ramos-Inza S, Ruberte AC, Sanmartín C, Sharma AK, Plano D. NSAIDs: Old Acquaintance in the Pipeline for Cancer Treatment and Prevention─Structural Modulation, Mechanisms of Action, and Bright Future. J Med Chem 2021; 64:16380-16421. [PMID: 34784195 DOI: 10.1021/acs.jmedchem.1c01460] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The limitations of current chemotherapeutic drugs are still a major issue in cancer treatment. Thus, targeted multimodal therapeutic approaches need to be strategically developed to successfully control tumor growth and prevent metastatic burden. Inflammation has long been recognized as a hallmark of cancer and plays a key role in the tumorigenesis and progression of the disease. Several epidemiological, clinical, and preclinical studies have shown that traditional nonsteroidal anti-inflammatory drugs (NSAIDs) exhibit anticancer activities. This Perspective reports the most recent outcomes for the treatment and prevention of different types of cancers for several NSAIDs alone or in combination with current chemotherapeutic drugs. Furthermore, an extensive review of the most promising structural modifications is reported, such as phospho, H2S, and NO releasing-, selenium-, metal complex-, and natural product-NSAIDs, among others. We also provide a perspective about the new strategies used to obtain more efficient NSAID- or NSAID derivative- formulations for targeted delivery.
Collapse
Affiliation(s)
- Sandra Ramos-Inza
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ana Carolina Ruberte
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| |
Collapse
|
20
|
Abu-Serie MM, Andrade F, Cámara-Sánchez P, Seras-Franzoso J, Rafael D, Díaz-Riascos ZV, Gener P, Abasolo I, Schwartz S. Pluronic F127 micelles improve the stability and enhance the anticancer stem cell efficacy of citral in breast cancer. Nanomedicine (Lond) 2021; 16:1471-1485. [PMID: 34160295 DOI: 10.2217/nnm-2021-0013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aim: Improving the stability and anti-cancer stem cell (CSC) activity of citral, a natural ALDH1A inhibitor. Materials & methods: Citral-loaded micelles (CLM) were obtained using Pluronic® F127 and its efficacy tested on the growth of four breast cancer cell lines. The impact of the CLM on the growth and functional hallmarks of breast CSCs were also evaluated using mammosphere and CSC reporter cell lines. Results: CLM improved the stability and growth inhibitory effects of citral. Importantly, CLM fully blocking the stemness features of CSCs (self-renewal, differentiation and migration) and in combination with paclitaxel CLM sensitized breast cancer cells to the chemotherapy. Conclusion: Targeting CSCs with CLM could improve the treatment of advanced breast cancer in combination with the standard chemotherapy.
Collapse
Affiliation(s)
- Marwa M Abu-Serie
- Department of Medical Biotechnology, Genetic Engineering, & Biotechnology Research Institute, City of Scientific Research & Technological Applications (SRTA-City), New Borg EL-Arab, 21934, Alexandria, Egypt
| | - Fernanda Andrade
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| | - Patricia Cámara-Sánchez
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain.,Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Joaquin Seras-Franzoso
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| | - Diana Rafael
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| | - Zamira V Díaz-Riascos
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain.,Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Petra Gener
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| | - Ibane Abasolo
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain.,Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
| | - Simó Schwartz
- Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 08035, Barcelona, Spain
| |
Collapse
|