1
|
Sapino S, Peira E, Chirio D, Chindamo G, Accomasso G, Vercelli C, Riganti C, Salaroglio IC, Gambino G, Re G, Amadori M, Gallarate M. Human and canine osteosarcoma cell lines: How do they react upon incubation with calcium phosphate-coated lipid nanoparticles carrying doxorubicin and curcumin? Int J Pharm 2025; 668:124970. [PMID: 39566701 DOI: 10.1016/j.ijpharm.2024.124970] [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: 07/25/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Osteosarcoma (OSA) is a bone cancer that affects both humans and animals, with dogs being particularly vulnerable. Standard therapy is often limited by multidrug resistance (MDR), primarily due to the overexpression of P-glycoprotein (P-gp), which expels drugs from the cells, reducing their efficacy. To overcome this challenge, drug delivery systems (DDS) and P-gp modulators have been explored. However, developing DDS that selectively target cancer cells remains difficult, with many current options lacking efficiency. Our research group has recently developed an innovative type of nanoparticle with a lipid core and a calcium phosphate shell (CaP-NPs), which enhances the uptake of doxorubicin (DOXO) in OSA cells. In this study, we loaded a lipophilic ester of doxorubicin (C12DOXO) and curcumin (CURC), a natural P-gp modulator, into CaP-NPs and co-incubated them into human and canine OSA cell lines, including DOXO-resistant cells. The results demonstrated a significant reduction in viability in human OSA cells. Additionally, the combination treatment led to a further increase in C12DOXO retention when cells were also treated with the P-gp inhibitor verapamil, indicating enhanced efficacy against MDR mechanisms. Notably, canine OSA cells exhibited a distinct response pattern, suggesting the presence of species-specific differences that warrant further investigation.
Collapse
Affiliation(s)
- Simona Sapino
- Department of Drug Science and Technology, Turin University, Via P. Giuria 9, 10125 Torino, Italy.
| | - Elena Peira
- Department of Drug Science and Technology, Turin University, Via P. Giuria 9, 10125 Torino, Italy.
| | - Daniela Chirio
- Department of Drug Science and Technology, Turin University, Via P. Giuria 9, 10125 Torino, Italy.
| | - Giulia Chindamo
- Department of Drug Science and Technology, Turin University, Via P. Giuria 9, 10125 Torino, Italy.
| | - Giulia Accomasso
- Department of Drug Science and Technology, Turin University, Via P. Giuria 9, 10125 Torino, Italy.
| | - Cristina Vercelli
- Department of Veterinary Sciences, Turin University, Largo Paolo Braccini 2, 10095 Grugliasco, Torino, Italy.
| | - Chiara Riganti
- Department of Oncology, Turin University, Piazza Nizza 44, 10126, Torino, Italy.
| | | | - Graziana Gambino
- Department of Veterinary Sciences, Turin University, Largo Paolo Braccini 2, 10095 Grugliasco, Torino, Italy.
| | - Giovanni Re
- Department of Veterinary Sciences, Turin University, Largo Paolo Braccini 2, 10095 Grugliasco, Torino, Italy.
| | - Michela Amadori
- Department of Veterinary Sciences, Turin University, Largo Paolo Braccini 2, 10095 Grugliasco, Torino, Italy.
| | - Marina Gallarate
- Department of Drug Science and Technology, Turin University, Via P. Giuria 9, 10125 Torino, Italy.
| |
Collapse
|
2
|
Kana Veedu A, Panthalattu Parambil A, Manheri MK. Sequential Release of Ibuprofen and the Gasotransmitter Hydrogen sulfide using Oxanorbornane-Based Synthetic Lipids as Carriers. Chempluschem 2024; 89:e202400323. [PMID: 39235160 DOI: 10.1002/cplu.202400323] [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: 05/06/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
After understanding the biological signaling roles of hydrogen sulfide and its involvement in various physiological processes, there has been enormous interest in exploring its therapeutic utility in areas such as cancer, inflammation, cardiovascular diseases, etc. There is also growing interest in using suitable H2S donors in combination with other drugs to improve the treatment outcome through the modulation of multiple pathways. The premature release of H2S from small molecule donors and the difficulty in controlling its spatio-temporal distribution are the major challenges during these efforts. Hence the development of appropriate carriers that can release this gasotransmitter along with the therapeutic entity of interest in a controlled manner has high significance. In this regard, this report presents a novel drug delivery system from oxanorbornane-based synthetic lipids that carries a H2S-releasing 1,2-dithiole-3-thione moiety as part of the head group. Nanoaggregates of the resulting conjugate are not only capable of efficiently entrapping a non-steroidal anti-inflammatory drug such as ibuprofen, but also release this drug and H2S in a controlled and sequential manner.
Collapse
Affiliation(s)
- Akshaya Kana Veedu
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamil Nadu, India
| | | | - Muraleedharan K Manheri
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamil Nadu, India
| |
Collapse
|
3
|
Jin Y, Yuan H, Liu Y, Zhu Y, Wang Y, Liang X, Gao W, Ren Z, Ji X, Wu D. Role of hydrogen sulfide in health and disease. MedComm (Beijing) 2024; 5:e661. [PMID: 39156767 PMCID: PMC11329756 DOI: 10.1002/mco2.661] [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: 11/04/2023] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 08/20/2024] Open
Abstract
In the past, hydrogen sulfide (H2S) was recognized as a toxic and dangerous gas; in recent years, with increased research, we have discovered that H2S can act as an endogenous regulatory transmitter. In mammals, H2S-catalyzing enzymes, such as cystathionine-β-synthase, cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase, are differentially expressed in a variety of tissues and affect a variety of biological functions, such as transcriptional and posttranslational modification of genes, activation of signaling pathways in the cell, and metabolic processes in tissues, by producing H2S. Various preclinical studies have shown that H2S affects physiological and pathological processes in the body. However, a detailed systematic summary of these roles in health and disease is lacking. Therefore, this review provides a thorough overview of the physiological roles of H2S in different systems and the diseases associated with disorders of H2S metabolism, such as ischemia-reperfusion injury, hypertension, neurodegenerative diseases, inflammatory bowel disease, and cancer. Meanwhile, this paper also introduces H2S donors and novel release modes, as well as the latest preclinical experimental results, aiming to provide researchers with new ideas to discover new diagnostic targets and therapeutic options.
Collapse
Affiliation(s)
- Yu‐Qing Jin
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Hang Yuan
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Ya‐Fang Liu
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Yi‐Wen Zhu
- School of Clinical MedicineHenan UniversityKaifengHenanChina
| | - Yan Wang
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Xiao‐Yi Liang
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Wei Gao
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Zhi‐Guang Ren
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Xin‐Ying Ji
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
- Faculty of Basic Medical SubjectsShu‐Qing Medical College of ZhengzhouZhengzhouHenanChina
| | - Dong‐Dong Wu
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
- School of StomatologyHenan UniversityKaifengHenanChina
- Department of StomatologyHuaihe Hospital of Henan UniversityKaifengHenanChina
| |
Collapse
|
4
|
Hanc K, Janeková H, Štacko P. Concurrent Subcellular Delivery of Hydrogen Sulfide and a Payload with Near-Infrared Light. JACS AU 2024; 4:2687-2694. [PMID: 39055161 PMCID: PMC11267537 DOI: 10.1021/jacsau.4c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024]
Abstract
Hydrogen sulfide (H2S) is a gaseous signaling molecule, exerting crucial regulatory functions in organelles and cellular environments. H2S exhibits high therapeutic potential and synergistic effects with other drugs, and its potency is notably enhanced through organelle-specific targeting. Yet, the navigation of light-activated H2S donors to specific organelles remains absent. Here, we report the first organelle-specific photocage that simultaneously delivers H2S and a payload with subcellular precision to mitochondria of live human cells using tissue-penetrating near-infrared light as a trigger. The fluorogenic payload enables real-time monitoring of the process, and we demonstrate the concurrent uncaging in mitochondria through a combination of fluorescence microscopy and mitochondria-specific fluorescent probes. We anticipate that these photocages will permit the precise delivery of H2S-drug combinations with exceptional spatiotemporal control, thereby driving the harnessing of known synergistic effects and the discovery of novel therapeutic strategies.
Collapse
Affiliation(s)
- Katarzyna Hanc
- Department of Chemistry, University
of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Hana Janeková
- Department of Chemistry, University
of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Peter Štacko
- Department of Chemistry, University
of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| |
Collapse
|
5
|
Xu Z. CRISPR/Cas9-mediated silencing of CD44: unveiling the role of hyaluronic acid-mediated interactions in cancer drug resistance. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2849-2876. [PMID: 37991544 DOI: 10.1007/s00210-023-02840-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023]
Abstract
A comprehensive overview of CD44 (CD44 Molecule (Indian Blood Group)), a cell surface glycoprotein, and its interaction with hyaluronic acid (HA) in drug resistance mechanisms across various types of cancer is provided, where CRISPR/Cas9 gene editing was utilized to silence CD44 expression and examine its impact on cancer cell behavior, migration, invasion, proliferation, and drug sensitivity. The significance of the HA-CD44 axis in tumor microenvironment (TME) delivery and its implications in specific cancer types, the influence of CD44 variants and the KHDRBS3 (KH RNA Binding Domain Containing, Signal Transduction Associated 3) gene on cancer progression and drug resistance, and the potential of targeting HA-mediated pathways using CRISPR/Cas9 gene editing technology to overcome drug resistance in cancer were also highlighted.
Collapse
Affiliation(s)
- Zhujun Xu
- Wuhan No.1 Hospital, Wuhan, 430022, Hubei, China.
| |
Collapse
|
6
|
Podolski-Renić A, Čipak Gašparović A, Valente A, López Ó, Bormio Nunes JH, Kowol CR, Heffeter P, Filipović NR. Schiff bases and their metal complexes to target and overcome (multidrug) resistance in cancer. Eur J Med Chem 2024; 270:116363. [PMID: 38593587 DOI: 10.1016/j.ejmech.2024.116363] [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: 02/27/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Overcoming multidrug resistance (MDR) is one of the major challenges in cancer therapy. In this respect, Schiff base-related compounds (bearing a R1R2CNR3 bond) gained high interest during the past decades. Schiff bases are considered privileged ligands for various reasons, including the easiness of their preparation and the possibility to form complexes with almost all transition metal ions. Schiff bases and their metal complexes exhibit many types of biological activities and are used for the treatment and diagnosis of various diseases. Until now, 13 Schiff bases have been investigated in clinical trials for cancer treatment and hypoxia imaging. This review represents the first collection of Schiff bases and their complexes which demonstrated MDR-reversal activity. The areas of drug resistance covered in this article involve: 1) Modulation of ABC transporter function, 2) Targeting lysosomal ABCB1 overexpression, 3) Circumvention of ABC transporter-mediated drug efflux by alternative routes of drug uptake, 4) Selective activity against MDR cancer models (collateral sensitivity), 5) Targeting GSH-detoxifying systems, 6) Overcoming apoptosis resistance by inducing necrosis and paraptosis, 7) Reactivation of mutated p53, 8) Restoration of sensitivity to DNA-damaging anticancer therapy, and 9) Overcoming drug resistance through modulation of the immune system. Through this approach, we would like to draw attention to Schiff bases and their metal complexes representing highly interesting anticancer drug candidates with the ability to overcome MDR.
Collapse
Affiliation(s)
- Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | | | - Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Óscar López
- Departamento de Química Organica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Julia H Bormio Nunes
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Nenad R Filipović
- Department of Chemistry and Biochemistry, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
7
|
Liu Y, Yue J, Ren Z, He M, Wang A, Xie J, Li T, Liu G, He X, Ge S, Yuan Y, Yang L. Vitamin C enhances the sensitivity of osteosarcoma to arsenic trioxide via inhibiting aerobic glycolysis. Toxicol Appl Pharmacol 2024; 482:116798. [PMID: 38160894 DOI: 10.1016/j.taap.2023.116798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
Osteosarcoma (OS) is a common malignant tumor disease in the department of orthopedics, which is prone to the age of adolescents and children under 20 years old. Arsenic trioxide (ATO), an ancient poison, has been reported to play a critical role in a variety of tumor treatments, including OS. However, due to certain poisonous side effects such as cardiotoxicity and hepatotoxicity, clinical application of ATO has been greatly limited. Here we report that low doses of ATO (1 μM) observably reduced the half-effective inhibitory concentration (IC50) of vitamin C on OS cells. Compared with the treatment alone, the synthetic application of vitamin C (VitC, 800 μM) and ATO (1 μM) significantly further inhibited the proliferation, migration, and invasion of OS cells and promoted cell apoptosis in vitro. Meanwhile, we observed that the combined application of VitC and ATO directly suppresses the aerobic glycolysis of OS cells with the decreased production of pyruvate, lactate, and ATP via inhibiting the expression of the critical glycolytic genes (PGK1, PGM1, and LDHA). Moreover, the combination of VitC (200 mg/kg) and ATO (1 mg/kg) with tail vein injection significantly delayed OS growth and migration of nude mice by inhibiting aerobic glycolysis of OS. Thus, our results demonstrate that VitC effectively increases the sensitivity of OS to low concentrations of ATO via inhibiting aerobic glycolysis to alleviate the toxic side effects of high doses of arsenic trioxide, suggesting that synthetic application of VitC and ATO is a promising approach for the clinical treatment of human OS.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jinrui Yue
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zijing Ren
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Mingyu He
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ao Wang
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jiajie Xie
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tao Li
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Guoxin Liu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xuting He
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Shiyu Ge
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ye Yuan
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China; National key laboratory of frigid cardiovascular disease, Harbin, China.
| | - Lei Yang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery of Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
8
|
Hou Y, Wang J, Wang J. Engineered biomaterial delivery strategies are used to reduce cardiotoxicity in osteosarcoma. Front Pharmacol 2023; 14:1284406. [PMID: 37854721 PMCID: PMC10579615 DOI: 10.3389/fphar.2023.1284406] [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: 08/28/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents. Chemotherapy drugs play an integral role in OS treatment. Preoperative neoadjuvant chemotherapy and postoperative conventional adjuvant chemotherapy improve survival in patients with OS. However, the toxic side effects of chemotherapy drugs are unavoidable. Cardiotoxicity is one of the common side effects of chemotherapy drugs that cannot be ignored. Chemotherapy drugs affect the destruction of mitochondrial autophagy and mitochondria-associated proteins to cause a decrease in cardiac ejection fraction and cardiomyocyte necrosis, which in turn causes heart failure and irreversible cardiomyopathy. Biomaterials play an important role in nanomedicine. Biomaterials act as carriers to deliver chemotherapy drugs precisely around tumor cells and continuously release carriers around the tumor. It not only promotes anti-tumor effects but also reduces the cardiotoxicity of chemotherapy drugs. In this paper, we first introduce the mechanism by which chemotherapy drugs commonly used in OS cause cardiotoxicity. Subsequently, we introduce biomaterials for reducing cardiotoxicity in OS chemotherapy. Finally, we prospect biomaterial delivery strategies to reduce cardiotoxicity in OS.
Collapse
Affiliation(s)
| | | | - Jianping Wang
- Department of Cardiology, Guangyuan Central Hospital, Guangyuan, China
| |
Collapse
|
9
|
Sodano F, Rolando B, Lazzarato L, Costamagna C, Failla M, Riganti C, Chegaev K. Use of Enzymatically Activated Carbon Monoxide Donors for Sensitizing Drug-Resistant Tumor Cells. Int J Mol Sci 2023; 24:11258. [PMID: 37511019 PMCID: PMC10379931 DOI: 10.3390/ijms241411258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
The application of gaseous signaling molecules like NO, H2S or CO to overcome the multidrug resistance in cancer treatment has proven to be a viable therapeutic strategy. The development of CO-releasing molecules (CORMs) in a controlled manner and in targeted tissues remains a challenge in medicinal chemistry. In this paper, we describe the design, synthesis and chemical and enzymatic stability of a novel non-metal CORM (1) able to release intracellularly CO and, simultaneously, facilitate fluorescent degradation of products under the action of esterase. The toxicity of 1 against different human cancer cell lines and their drug-resistant counterparts, as well as the putative mechanism of toxicity were investigated. The drug-resistant cancer cell lines efficiently absorbed 1 and 1 was able to restore their sensitivity vs. chemotherapeutic drugs by causing a CO-dependent mitochondrial oxidative stress that culminated in mitochondrial-dependent apoptosis. These results demonstrate the importance of CORMs in cases where conventional chemotherapy fails and thus open the horizons towards new combinatorial strategies to overcome multidrug resistance.
Collapse
Affiliation(s)
- Federica Sodano
- Department of Pharmacy, "Federico II" University of Naples, 80131 Naples, Italy
| | - Barbara Rolando
- Department of Drug Science and Technology, University of Torino, 10125 Torino, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Torino, 10125 Torino, Italy
| | | | - Mariacristina Failla
- Department of Drug Science and Technology, University of Torino, 10125 Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10125 Torino, Italy
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Torino, 10125 Torino, Italy
| |
Collapse
|
10
|
H2S Donors with Cytoprotective Effects in Models of MI/R Injury and Chemotherapy-Induced Cardiotoxicity. Antioxidants (Basel) 2023; 12:antiox12030650. [PMID: 36978898 PMCID: PMC10045576 DOI: 10.3390/antiox12030650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous signaling molecule that greatly influences several important (patho)physiological processes related to cardiovascular health and disease, including vasodilation, angiogenesis, inflammation, and cellular redox homeostasis. Consequently, H2S supplementation is an emerging area of interest, especially for the treatment of cardiovascular-related diseases. To fully unlock the medicinal properties of hydrogen sulfide, however, the development and refinement of H2S releasing compounds (or donors) are required to augment its bioavailability and to better mimic its natural enzymatic production. Categorizing donors by the biological stimulus that triggers their H2S release, this review highlights the fundamental chemistry and releasing mechanisms of a range of H2S donors that have exhibited promising protective effects in models of myocardial ischemia-reperfusion (MI/R) injury and cancer chemotherapy-induced cardiotoxicity, specifically. Thus, in addition to serving as important investigative tools that further advance our knowledge and understanding of H2S chemical biology, the compounds highlighted in this review have the potential to serve as vital therapeutic agents for the treatment (or prevention) of various cardiomyopathies.
Collapse
|
11
|
He B, Zhang Z, Huang Z, Duan X, Wang Y, Cao J, Li L, He K, Nice EC, He W, Gao W, Shen Z. Protein persulfidation: Rewiring the hydrogen sulfide signaling in cell stress response. Biochem Pharmacol 2023; 209:115444. [PMID: 36736962 DOI: 10.1016/j.bcp.2023.115444] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
The past few decades have witnessed significant progress in the discovery of hydrogen sulfide (H2S) as a ubiquitous gaseous signaling molecule in mammalian physiology, akin to nitric oxide and carbon monoxide. As the third gasotransmitter, H2S is now known to exert a wide range of physiological and cytoprotective functions in the biological systems. However, endogenous H2S concentrations are usually low, and its potential biologic mechanisms responsible have not yet been fully clarified. Recently, a growing body of evidence has demonstrated that protein persulfidation, a posttranslational modification of cysteine residues (RSH) to persulfides (RSSH) elicited by H2S, is a fundamental mechanism of H2S-mediated signaling pathways. Persulfidation, as a biological switch for protein function, plays an important role in the maintenance of cell homeostasis in response to various internal and external stress stimuli and is also implicated in numerous diseases, such as cardiovascular and neurodegenerative diseases and cancer. In this review, the biological significance of protein persulfidation by H2S in cell stress response is reviewed providing a framework for understanding the multifaceted roles of H2S. A mechanism-guided perspective can help open novel avenues for the exploitation of therapeutics based on H2S-induced persulfidation in the context of diseases.
Collapse
Affiliation(s)
- Bo He
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhe Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhao Huang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xirui Duan
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yu Wang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jiangjun Cao
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Kai He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Weifeng He
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Army Military Medical University, Chongqing 400038, China.
| | - Wei Gao
- Clinical Genetics Laboratory, Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu 610081, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China.
| |
Collapse
|
12
|
Hattinger CM, Salaroglio IC, Fantoni L, Godel M, Casotti C, Kopecka J, Scotlandi K, Ibrahim T, Riganti C, Serra M. Strategies to Overcome Resistance to Immune-Based Therapies in Osteosarcoma. Int J Mol Sci 2023; 24:ijms24010799. [PMID: 36614241 PMCID: PMC9821333 DOI: 10.3390/ijms24010799] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Improving the prognosis and cure rate of HGOSs (high-grade osteosarcomas) is an absolute need. Immune-based treatment approaches have been increasingly taken into consideration, in particular for metastatic, relapsed and refractory HGOS patients, to ameliorate the clinical results currently achieved. This review is intended to give an overview on the immunotherapeutic treatments targeting, counteracting or exploiting the different immune cell compartments that are present in the HGOS tumor microenvironment. The principle at the basis of these strategies and the possible mechanisms that HGOS cells may use to escape these treatments are presented and discussed. Finally, a list of the currently ongoing immune-based trials in HGOS is provided, together with the results that have been obtained in recently completed clinical studies. The different strategies that are presently under investigation, which are generally aimed at abrogating the immune evasion of HGOS cells, will hopefully help to indicate new treatment protocols, leading to an improvement in the prognosis of patients with this tumor.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | | | - Leonardo Fantoni
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Martina Godel
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Chiara Casotti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
- Correspondence: (C.R.); (M.S.)
| | - Massimo Serra
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Correspondence: (C.R.); (M.S.)
| |
Collapse
|
13
|
Rong F, Wang T, Zhou Q, Peng H, Yang J, Fan Q, Li P. Intelligent polymeric hydrogen sulfide delivery systems for therapeutic applications. Bioact Mater 2023; 19:198-216. [PMID: 35510171 PMCID: PMC9034248 DOI: 10.1016/j.bioactmat.2022.03.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 12/21/2022] Open
Abstract
Hydrogen sulfide (H2S) plays an important role in regulating various pathological processes such as protecting mammalian cell from harmful injuries, promoting tissue regeneration, and regulating the process of various diseases caused by physiological disorders. Studies have revealed that the physiological effects of H2S are highly associated with its concentrations. At relatively low concentration, H2S shows beneficial functions. However, long-time and high-dose donation of H2S would inhibit regular biological process, resulting in cell dysfunction and apoptosis. To regulate the dosage of H2S delivery for precision medicine, H2S delivery systems with intelligent characteristics were developed and a variety of biocompatibility polymers have been utilized to establish intelligent polymeric H2S delivery systems, with the abilities to specifically target the lesions, smartly respond to pathological microenvironments, as well as real-timely monitor H2S delivery and lesion conditions by incorporating imaging-capable moieties. In this review, we focus on the design, preparation, and therapeutic applications of intelligent polymeric H2S delivery systems in cardiovascular therapy, inflammatory therapy, tissue regenerative therapy, cancer therapy and bacteria-associated therapy. Strategies for precise H2S therapies especially imaging-guided H2S theranostics are highlighted. Since H2S donors with stimuli-responsive characters are vital components for establishing intelligent H2S delivery systems, the development of H2S donors is also briefly introduced. H2S is an endogenous gasotransmitter that plays important role in regulating various physiological and pathological pathways. Controlled H2S delivery is vital since the therapeutic effects of H2S are highly associated with its concentrations. Intelligent polymeric H2S delivery systems possess specific targeting, stimuli responsive and imaging guided capabilities, representing a strategic option for next generation of therapies.
Collapse
|
14
|
Zheng K, Bai J, Yang H, Xu Y, Pan G, Wang H, Geng D. Nanomaterial-assisted theranosis of bone diseases. Bioact Mater 2022; 24:263-312. [PMID: 36632509 PMCID: PMC9813540 DOI: 10.1016/j.bioactmat.2022.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/27/2022] Open
Abstract
Bone-related diseases refer to a group of skeletal disorders that are characterized by bone and cartilage destruction. Conventional approaches can regulate bone homeostasis to a certain extent. However, these therapies are still associated with some undesirable problems. Fortunately, recent advances in nanomaterials have provided unprecedented opportunities for diagnosis and therapy of bone-related diseases. This review provides a comprehensive and up-to-date overview of current advanced theranostic nanomaterials in bone-related diseases. First, the potential utility of nanomaterials for biological imaging and biomarker detection is illustrated. Second, nanomaterials serve as therapeutic delivery platforms with special functions for bone homeostasis regulation and cellular modulation are highlighted. Finally, perspectives in this field are offered, including current key bottlenecks and future directions, which may be helpful for exploiting nanomaterials with novel properties and unique functions. This review will provide scientific guidance to enhance the development of advanced nanomaterials for the diagnosis and therapy of bone-related diseases.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author.Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China,Corresponding author.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author. Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
15
|
Zhu P, Li T, Li Q, Gu Y, Shu Y, Hu K, Chen L, Peng X, Peng J, Hao L. Mechanism and Role of Endoplasmic Reticulum Stress in Osteosarcoma. Biomolecules 2022; 12:1882. [PMID: 36551309 PMCID: PMC9775044 DOI: 10.3390/biom12121882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor, often occurring in children and adolescents. The etiology of most patients is unclear, and the current conventional treatment methods are chemotherapy, radiotherapy, and surgical resection. However, the sensitivity of osteosarcoma to radiotherapy and chemotherapy is low, and the prognosis is poor. The development of new and useful treatment strategies for improving patient survival is an urgent need. It has been found that endoplasmic reticulum (ER) stress (ERS) affects tumor angiogenesis, invasion, etc. By summarizing the literature related to osteosarcoma and ERS, we found that the unfolded protein response (UPR) pathway activated by ERS has a regulatory role in osteosarcoma proliferation, apoptosis, and chemoresistance. In osteosarcoma, the UPR pathway plays an important role by crosstalk with autophagy, oxidative stress, and other pathways. Overall, this article focuses on the relationship between ERS and osteosarcoma and reviews the potential of drugs or gene targets associated with ERS for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Peijun Zhu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Ting Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qingqing Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yawen Gu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yuan Shu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Kaibo Hu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Leifeng Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaogang Peng
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jie Peng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liang Hao
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
16
|
Hydrogen Sulfide Attenuates Lipopolysaccharide-Induced Inflammation via the P-glycoprotein and NF-κB Pathway in Astrocytes. Neurochem Res 2022; 48:1424-1437. [PMID: 36482035 PMCID: PMC10066098 DOI: 10.1007/s11064-022-03840-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/13/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
Abstract
AbstractAstrocyte activation is key in neurodegenerative diseases. Hydrogen sulfide (H2S) exhibits neuroprotective effects on astrocytes, although the underlying molecular mechanism remains unclear. Here, we explored the effects of H2S on lipopolysaccharide (LPS)-induced astrocyte activation and astrocyte-mediated neuroinflammation. After inducing primary astrocytes via LPS exposure, H2S levels were altered. The generation and secretion of inflammatory mediators by astrocytes and their interrelation with P-glycoprotein (P-gp), an important transporter belonging to the ABC transporter family, were assessed. Activated astrocytes showed upregulated glial fibrillary acidic protein (GFAP) mRNA expression, and significantly increased proinflammatory factor mRNA/protein expression and release. The secretory capacity of astrocytes was reduced, with significantly decreased proinflammatory factor levels in culture supernatant after P-gp inhibitor verapamil pretreatment. The increase in the intracellular H2S level inhibited LPS-induced GFAP expression and P65 nuclear entry in astrocytes. mRNA expression and release of proinflammatory factors were reduced significantly, with no significant changes in cytoplasmic protein expression. S-sulfhydration levels increased significantly with the increased concentration of sodium hydrosulfide or S-adenosyl-l-methionine addition, with only moderate changes in astrocyte P-gp expression. H2S regulates NF-κB activation, leads to S-sulfhydration of P-gp, and inhibits the biosynthesis and secretion of proinflammatory factors by astrocytes. The regulatory effects of H2S on astrocytes may have clinical value for exploring new therapeutic strategies against neurodegenerative diseases.
Collapse
|
17
|
On-demand therapeutic delivery of hydrogen sulfide aided by biomolecules. J Control Release 2022; 352:586-599. [PMID: 36328076 DOI: 10.1016/j.jconrel.2022.10.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
Hydrogen sulfide (H2S), known as the third gasotransmitter, exerts various physiological functions including cardiac protection, angiogenesis, anti-inflammatory, and anti-cancer capability. Given its promising therapeutic potential as well as severe perniciousness if improper use, the sustained and tunable H2S delivery systems are highly required for H2S-based gas therapy with enhanced bioactivity and reduced side effects. To this end, a series of stimuli-responsive compounds capable of releasing H2S (termed H2S donors) have been designed over the past two decades to mimic the endogenous generation of H2S and elucidate the biological functions. Further to improve the stability of H2S donors and achieve the targeted delivery, various delivery systems have been constructed. In this review, we focus on the recent advances of an emerging subset, biomolecular-based H2S delivery systems, which combine H2S donors with biomolecular vectors including polysaccharide, peptide, and protein. We demonstrated their basic structures, building strategies, and therapeutic applications respectively to unfold their inherent merits endued by biomolecules including biocompatibility, biodegradability as well as expansibility. The varied development potentials of biomolecular-based H2S delivery systems based on their specific properties are also discussed. At the end, brief future outlooks and upcoming challenges are presented as well.
Collapse
|
18
|
Yang C, Li X, Yan Q. Polythionoester Vesicle: An Efficient Polymeric Platform for Tuning H 2S Release. ACS Macro Lett 2022; 11:1230-1237. [PMID: 36223277 DOI: 10.1021/acsmacrolett.2c00473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hydrogen sulfide (H2S) serves as a key gaseous regulator that not only directs many physiological activities, but also manifests therapeutic benefits to many diseases. Developing H2S vehicle platforms for its local delivery and long-acting release is important to achieve target gas therapy. Most of the known H2S-donating polymers contain labile thioester scaffolds within their structures that suffer from the issue of low gas releasing efficiency. Here we present the use of thionoester, a constitutional isomer of thioester, as the functional unit to build a structural platform of cysteine-triggered H2S donor polymer, polythionoester. Simple exchange of the sulfur and oxygen positions in the carbonyl sulfide scaffold makes the polythionoesters undergo a distinct mechanism of H2S production, which can largely improve the gas-releasing efficiency (>80%). Moreover, the thionoester-containing block copolymers can self-assemble into vesicles in an aqueous media. We discover that control over the size effect can adjust the vesicle disassembly rate and gas-releasing kinetics. A tunable half-life of H2S generation (2.6-9.8 h) can be accessed by tailoring the vesicle dimension. This allows such polymersomes to be potential as a gas nanodelivery system for long-lasting gas therapeutics.
Collapse
Affiliation(s)
- Cuiqin Yang
- State Key Lab of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Xuefeng Li
- State Key Lab of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Qiang Yan
- State Key Lab of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| |
Collapse
|
19
|
Naked and Decorated Nanoparticles Containing H 2S-Releasing Doxorubicin: Preparation, Characterization and Assessment of Their Antitumoral Efficiency on Various Resistant Tumor Cells. Int J Mol Sci 2022; 23:ijms231911555. [PMID: 36232858 PMCID: PMC9569649 DOI: 10.3390/ijms231911555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Several semisynthetic, low-cardiotoxicity doxorubicin (DOXO) conjugated have been extensively described, considering the risk of cytotoxicity loss against resistant tumor cells, which mainly present drug efflux capacity. Doxorubicin 14-[4-(4-phenyl-5-thioxo-5H-[1,2]dithiol-3-yl)]-benzoate (H2S-DOXO) was synthetized and tested for its ability to overcome drug resistance with good intracellular accumulation. In this paper, we present a formulation study aimed to develop naked and decorated H2S-DOXO-loaded lipid nanoparticles (NPs). NPs prepared by the "cold dilution of microemulsion" method were decorated with hyaluronic acid (HA) to obtain active targeting and characterized for their physicochemical properties, drug entrapment efficiency, long-term stability, and in vitro drug release. Best formulations were tested in vitro on human-sensitive (MCF7) and human/mouse DOXO-resistant (MDA-MDB -231 and JC) breast cancer cells, on human (U-2OS) osteosarcoma cells and DOXO-resistant human/mouse osteosarcoma cells (U-2OS/DX580/K7M2). HA-decoration by HA-cetyltrimethyl ammonium bromide electrostatic interaction on NPs surface was confirmed by Zeta potential and elemental analysis at TEM. NPs had mean diameters lower than 300 nm, 70% H2S-DOXO entrapment efficiency, and were stable for almost 28 days. HA-decorated NPs accumulated H2S-DOXO in Pgp-expressing cells reducing cell viability. HA-decorated NPs result in the best formulation to increase the inter-cellular H2S-DOXO delivery and kill resistant cells, and therefore, as a future perspective, they will be taken into account for further in vivo experiments on tumor animal model.
Collapse
|
20
|
IL-11Rα-targeted nanostrategy empowers chemotherapy of relapsed and patient-derived osteosarcoma. J Control Release 2022; 350:460-470. [PMID: 36041590 DOI: 10.1016/j.jconrel.2022.08.048] [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: 07/18/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022]
Abstract
Osteosarcoma (OS) is a rare but frequently lethal bone malignancy in children and adolescents. The adjuvant chemotherapy with doxorubicin (Dox) and cisplatin remains a mainstream clinical practice though it affords only limited clinical benefits due to low tumor deposition, dose-limiting toxicity and high rate of relapse/metastasis. Here, taking advantage of high IL-11Rα expression in the OS patients, we installed IL-11Rα specific peptide (sequence: CGRRAGGSC) onto redox-responsive polymersomes encapsulating Dox (IL11-PDox) to boost the specificity and anti-OS efficacy of chemotherapy. Of note, IL-11Rα peptide at a density of 20% greatly augmented the internalization, apoptotic activity, and migration inhibition of Dox in IL-11Rα-overexpressing 143B OS cells. The active targeting effect of IL-11-PDox was supported in orthotopic and relapsed 143B OS models, as shown by striking repression of tumor growth and lung metastasis and substantial survival benefits over free Dox control. We further verified that IL11-PDox could effectively inhibit patient-derived OS xenografts. IL-11Rα-targeted nanodelivery of chemotherapeutics provides a potential therapeutic strategy for advanced osteosarcoma.
Collapse
|
21
|
Fernández-Tabanera E, Melero-Fernández de Mera RM, Alonso J. CD44 In Sarcomas: A Comprehensive Review and Future Perspectives. Front Oncol 2022; 12:909450. [PMID: 35785191 PMCID: PMC9247467 DOI: 10.3389/fonc.2022.909450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 12/16/2022] Open
Abstract
It is widely accepted that the tumor microenvironment, particularly the extracellular matrix, plays an essential role in the development of tumors through the interaction with specific protein-membrane receptors. One of the most relevant proteins in this context is the transmembrane protein CD44. The role of CD44 in tumor progression, invasion, and metastasis has been well established in many cancers, although a comprehensive review concerning its role in sarcomas has not been published. CD44 is overexpressed in most sarcomas and several in vitro and in vivo experiments have shown a direct effect on tumor progression, dissemination, and drug resistance. Moreover, CD44 has been revealed as a useful marker for prognostic and diagnostic (CD44v6 isoform) in osteosarcoma. Besides, some innovative treatments such as HA-functionalized liposomes therapy have become an excellent CD44-mediated intracellular delivery system for osteosarcoma. Unfortunately, the reduced number of studies deciphering the prognostic/diagnostic value of CD44 in other sarcoma subgroups, neither than osteosarcoma, in addition to the low number of patients involved in those studies, have produced inconclusive results. In this review, we have gone through the information available on the role of CD44 in the development, maintenance, and progression of sarcomas, analyzing their implications at the prognostic, therapeutic, and mechanistic levels. Moreover, we illustrate how research involving the specific role of CD44 in the different sarcoma subgroups could suppose a chance to advance towards a more innovative perspective for novel therapies and future clinical trials.
Collapse
Affiliation(s)
- Enrique Fernández-Tabanera
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
- Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Raquel M. Melero-Fernández de Mera
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
| | - Javier Alonso
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
- *Correspondence: Javier Alonso,
| |
Collapse
|
22
|
Alov P, Al Sharif M, Aluani D, Chegaev K, Dinic J, Divac Rankov A, Fernandes MX, Fusi F, García-Sosa AT, Juvonen R, Kondeva-Burdina M, Padrón JM, Pajeva I, Pencheva T, Puerta A, Raunio H, Riganti C, Tsakovska I, Tzankova V, Yordanov Y, Saponara S. A Comprehensive Evaluation of Sdox, a Promising H2S-Releasing Doxorubicin for the Treatment of Chemoresistant Tumors. Front Pharmacol 2022; 13:831791. [PMID: 35321325 PMCID: PMC8936434 DOI: 10.3389/fphar.2022.831791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Sdox is a hydrogen sulfide (H2S)-releasing doxorubicin effective in P-glycoprotein-overexpressing/doxorubicin-resistant tumor models and not cytotoxic, as the parental drug, in H9c2 cardiomyocytes. The aim of this study was the assessment of Sdox drug-like features and its absorption, distribution, metabolism, and excretion (ADME)/toxicity properties, by a multi- and transdisciplinary in silico, in vitro, and in vivo approach. Doxorubicin was used as the reference compound. The in silico profiling suggested that Sdox possesses higher lipophilicity and lower solubility compared to doxorubicin, and the off-targets prediction revealed relevant differences between Dox and Sdox towards several cancer targets, suggesting different toxicological profiles. In vitro data showed that Sdox is a substrate with lower affinity for P-glycoprotein, less hepatotoxic, and causes less oxidative damage than doxorubicin. Both anthracyclines inhibited CYP3A4, but not hERG currents. Unlike doxorubicin, the percentage of zebrafish live embryos at 72 hpf was not affected by Sdox treatment. In conclusion, these findings demonstrate that Sdox displays a more favorable drug-like ADME/toxicity profile than doxorubicin, different selectivity towards cancer targets, along with a greater preclinical efficacy in resistant tumors. Therefore, Sdox represents a prototype of innovative anthracyclines, worthy of further investigations in clinical settings.
Collapse
Affiliation(s)
- Petko Alov
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Merilin Al Sharif
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Denitsa Aluani
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Jelena Dinic
- Department of Neurobiology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Divac Rankov
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Miguel X. Fernandes
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Risto Juvonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Magdalena Kondeva-Burdina
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - José M. Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Ilza Pajeva
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tania Pencheva
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Hannu Raunio
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | - Ivanka Tsakovska
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Virginia Tzankova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Yordan Yordanov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Siena, Italy
- *Correspondence: Simona Saponara,
| |
Collapse
|
23
|
Durante M, Frosini M, Chiaino E, Fusi F, Gamberucci A, Gorelli B, Chegaev K, Riganti C, Saponara S. Sdox, a H 2S releasing anthracycline, with a safer profile than doxorubicin toward vasculature. Vascul Pharmacol 2022; 143:106969. [PMID: 35149209 DOI: 10.1016/j.vph.2022.106969] [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: 09/15/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 01/18/2023]
Abstract
Sdox is a synthetic H2S-releasing doxorubicin (Dox) less cardiotoxic and more effective than Dox in pre-clinical, Dox-resistant tumour models. The well-known anthracycline vascular toxicity, however, might limit Sdox clinical use. This study aimed at evaluating Sdox vascular toxicity in vitro, using Dox as reference compound. Both vascular smooth muscle A7r5 and endothelial EA.hy926 cells were more sensitive to Dox than Sdox, although both drugs equally increased intracellular free radical levels. Sdox released H2S in both cell lines. The H2S scavenger hydroxocobalamin partially reverted Sdox-induced cytotoxicity in A7r5, but not in EA.hy926 cells, suggesting a role for H2S in smooth muscle cell death. Markers of Sdox-induced apoptosis were significantly lower than, in A7r5 cells, and comparable to those of Dox in EA.hy926 cells. In A7r5 cells, Dox increased the activity of caspase 3, 8, and 9, Sdox affecting only that of caspase 3. Moreover, both drugs induced comparable DNA damage in A7r5 cells, while Sdox was less toxic than Dox in Ea.hy926 cells. In fresh aorta rings, only Dox weakly increased phenylephrine-induced contraction when endothelium was present. In rings cultured with both drugs for 7 days, Sdox blunted phenylephrine- and high K+-induced contractions though at a concentration 10-fold higher than that of Dox. In conclusion, Sdox may represent the prototype of an innovative anthracycline, effective against Dox-resistant tumours, displaying a more favourable vascular toxicity profile compared to the parent compound.
Collapse
Affiliation(s)
- Miriam Durante
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Maria Frosini
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Elda Chiaino
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Fabio Fusi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Alessandra Gamberucci
- Dipartimento di Medicina Molecolare e dello Sviluppo, Università di Siena, Siena, Italy
| | - Beatrice Gorelli
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Konstantin Chegaev
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, Torino, Italy
| | - Chiara Riganti
- Dipartimento di Oncologia, Università di Torino, Torino, Italy
| | - Simona Saponara
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy.
| |
Collapse
|
24
|
Hu J, Fang Y, Huang X, Qiao R, Quinn JF, Davis TP. Engineering macromolecular nanocarriers for local delivery of gaseous signaling molecules. Adv Drug Deliv Rev 2021; 179:114005. [PMID: 34687822 DOI: 10.1016/j.addr.2021.114005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/30/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023]
Abstract
In addition to being notorious air pollutants, nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) have also been known as endogenous gaseous signaling molecules (GSMs). These GSMs play critical roles in maintaining the homeostasis of living organisms. Importantly, the occurrence and development of many diseases such as inflammation and cancer are highly associated with the concentration changes of GSMs. As such, GSMs could also be used as new therapeutic agents, showing great potential in the treatment of many formidable diseases. Although clinically it is possible to directly inhale GSMs, the precise control of the dose and concentration for local delivery of GSMs remains a substantial challenge. The development of gaseous signaling molecule-releasing molecules provides a great tool for the safe and convenient delivery of GSMs. In this review article, we primarily focus on the recent development of macromolecular nanocarriers for the local delivery of various GSMs. Learning from the chemistry of small molecule-based donors, the integration of these gaseous signaling molecule-releasing molecules into polymeric matrices through physical encapsulation, post-modification, or direct polymerization approach renders it possible to fabricate numerous macromolecular nanocarriers with optimized pharmacokinetics and pharmacodynamics, revealing improved therapeutic performance than the small molecule analogs. The development of GSMs represents a new means for many disease treatments with unique therapeutic outcomes.
Collapse
|
25
|
Advances in understanding the role of P-gp in doxorubicin resistance: Molecular pathways, therapeutic strategies, and prospects. Drug Discov Today 2021; 27:436-455. [PMID: 34624510 DOI: 10.1016/j.drudis.2021.09.020] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/22/2021] [Accepted: 09/29/2021] [Indexed: 12/11/2022]
Abstract
P-glycoprotein (P-gp) is a drug efflux transporter that triggers doxorubicin (DOX) resistance. In this review, we highlight the molecular avenues regulating P-gp, such as Nrf2, HIF-1α, miRNAs, and long noncoding (lnc)RNAs, to reveal their participation in DOX resistance. These antitumor compounds and genetic tools synergistically reduce P-gp expression. Furthermore, ATP depletion impairs P-gp activity to enhance the antitumor activity of DOX. Nanoarchitectures, including liposomes, micelles, polymeric nanoparticles (NPs), and solid lipid nanocarriers, have been developed for the co-delivery of DOX with anticancer compounds and genes enhancing DOX cytotoxicity. Surface modification of nanocarriers, for instance with hyaluronic acid (HA), can promote selectivity toward cancer cells. We discuss these aspects with a focus on P-gp expression and activity.
Collapse
|
26
|
Argenziano M, Arpicco S, Brusa P, Cavalli R, Chirio D, Dosio F, Gallarate M, Peira E, Stella B, Ugazio E. Developing Actively Targeted Nanoparticles to Fight Cancer: Focus on Italian Research. Pharmaceutics 2021; 13:pharmaceutics13101538. [PMID: 34683830 PMCID: PMC8540327 DOI: 10.3390/pharmaceutics13101538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 01/02/2023] Open
Abstract
Active targeting is a valuable and promising approach with which to enhance the therapeutic efficacy of nanodelivery systems, and the development of tumor-targeted nanoparticles has therefore attracted much research attention. In this field, the research carried out in Italian Pharmaceutical Technology academic groups has been focused on the development of actively targeted nanosystems using a multidisciplinary approach. To highlight these efforts, this review reports a thorough description of the last 10 years of Italian research results on the development of actively targeted nanoparticles to direct drugs towards different receptors that are overexpressed on cancer cells or in the tumor microenvironment. In particular, the review discusses polymeric nanocarriers, liposomes, lipoplexes, niosomes, solid lipid nanoparticles, squalene nanoassemblies and nanobubbles. For each nanocarrier, the main ligands, conjugation strategies and target receptors are described. The literature indicates that polymeric nanoparticles and liposomes stand out as key tools for improving specific drug delivery to the site of action. In addition, solid lipid nanoparticles, squalene nanoparticles and nanobubbles have also been successfully proposed. Taken together, these strategies all offer many platforms for the design of nanocarriers that are suitable for future clinical translation.
Collapse
Affiliation(s)
| | - Silvia Arpicco
- Correspondence: (S.A.); (M.G.); Tel.: +39-011-670-6668 (S.A.); +39-011-670-7194 (M.G.)
| | | | | | | | | | - Marina Gallarate
- Correspondence: (S.A.); (M.G.); Tel.: +39-011-670-6668 (S.A.); +39-011-670-7194 (M.G.)
| | | | | | | |
Collapse
|
27
|
Drug Resistance in Osteosarcoma: Emerging Biomarkers, Therapeutic Targets and Treatment Strategies. Cancers (Basel) 2021; 13:cancers13122878. [PMID: 34207685 PMCID: PMC8228414 DOI: 10.3390/cancers13122878] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/05/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Despite the adoption of aggressive, multimodal treatment schedules, the cure rate of high-grade osteosarcoma (HGOS) has not significantly improved in the last 30 years. The most relevant problem preventing improvement in HGOS prognosis is drug resistance. Therefore, validated novel biomarkers that help to identify those patients who could benefit from innovative treatment options and the development of drugs enabling personalized therapeutic protocols are necessary. The aim of this review was to give an overview on the most relevant emerging drug resistance-related biomarkers, therapeutic targets and new agents or novel candidate treatment strategies, which have been highlighted and suggested for HGOS to improve the success rate of clinical trials. Abstract High-grade osteosarcoma (HGOS), the most common primary malignant tumor of bone, is a highly aggressive neoplasm with a cure rate of approximately 40–50% in unselected patient populations. The major clinical problems opposing the cure of HGOS are the presence of inherent or acquired drug resistance and the development of metastasis. Since the drugs used in first-line chemotherapy protocols for HGOS and clinical outcome have not significantly evolved in the past three decades, there is an urgent need for new therapeutic biomarkers and targeted treatment strategies, which may increase the currently available spectrum of cure modalities. Unresponsive or chemoresistant (refractory) HGOS patients usually encounter a dismal prognosis, mostly because therapeutic options and drugs effective for rescue treatments are scarce. Tailored treatments for different subgroups of HGOS patients stratified according to drug resistance-related biomarkers thus appear as an option that may improve this situation. This review explores drug resistance-related biomarkers, therapeutic targets and new candidate treatment strategies, which have emerged in HGOS. In addition to consolidated biomarkers, specific attention has been paid to the role of non-coding RNAs, tumor-derived extracellular vesicles, and cancer stem cells as contributors to drug resistance in HGOS, in order to highlight new candidate markers and therapeutic targets. The possible use of new non-conventional drugs to overcome the main mechanisms of drug resistance in HGOS are finally discussed.
Collapse
|
28
|
Liao YX, Lv JY, Zhou ZF, Xu TY, Yang D, Gao QM, Fan L, Li GD, Yu HY, Liu KY. CXCR4 blockade sensitizes osteosarcoma to doxorubicin by inducing autophagic cell death via PI3K‑Akt‑mTOR pathway inhibition. Int J Oncol 2021; 59:49. [PMID: 34080667 PMCID: PMC8208619 DOI: 10.3892/ijo.2021.5229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022] Open
Abstract
Doxorubicin is one of the most frequently used chemotherapy drugs in the treatment of osteosarcoma (OS), but the emergence of chemoresistance often leads to treatment failure. C-X-C motif chemokine receptor 4 (CXCR4) has been demonstrated to regulate OS progression and metastasis. However, whether CXCR4 is also involved in OS chemoresistance and its molecular mechanisms has yet to be fully elucidated. In the present study, CXCR4-mediated autophagy for OS chemotherapy was investigated by western blot analysis, transmission electron microscopy and confocal microscopy. CXCR4 silencing enhanced doxorubicin-induced apoptosis by reducing P-glycoprotein in CXCR4+ LM8 cells, while CXCR4 overexpression promoted OS doxorubicin resistance in CXCR4−Dunn cells. Furthermore, CXCR4 silencing with or without doxorubicin increased the expression of beclin 1 and light chain 3B, and the number of autophagosomes and autolysosomes, as well as induced autophagic flux activation by suppressing the PI3K/AKT/mTOR signaling pathway. In addition, pretreatment with the autophagy inhibitor bafilomycin A1 attenuated CXCR4 abrogation-induced cell death. Finally, the CXCR4 antagonist AMD3100 synergistically reinforced the antitumor effect of doxorubicin in an orthotopic OS mouse model. Taken together, the present study revealed that CXCR4 inhibition sensitizes OS to doxorubicin by inducing autophagic cell death. Therefore, targeting the CXCR4/autophagy axis may be a promising therapeutic strategy to overcome OS chemotherapy resistance.
Collapse
Affiliation(s)
- Yu-Xin Liao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Ji-Yang Lv
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Zi-Fei Zhou
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Tian-Yang Xu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Dong Yang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Qiu-Ming Gao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Lin Fan
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Guo-Dong Li
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Hai-Yang Yu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Kai-Yuan Liu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
29
|
Zhang S, Guo W. β-Elemene Enhances the Sensitivity of Osteosarcoma Cells to Doxorubicin via Downregulation of Peroxiredoxin-1. Onco Targets Ther 2021; 14:3599-3609. [PMID: 34113126 PMCID: PMC8184248 DOI: 10.2147/ott.s303152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
Background Doxorubicin (Dox) resistance is a primary obstacle for the treatment of osteosarcoma. Meanwhile, β-Elemene was shown to exhibit an anti-proliferative effect on osteosarcoma cells. However, the role of a combination of Dox with β-Elemene on osteosarcoma cells remains unclear. Thus, this study aimed to investigate the role of the combination of Dox with β-Elemene on the proliferation, apoptosis and oxidative stress of Dox-resistance osteosarcoma cells. Methods CKC-8, EdU staining and flow cytometry assays were used to determine the viability, proliferation and apoptosis of Dox-resistance osteosarcoma cells, respectively. Meanwhile, the expression of antioxidant protein peroxiredoxin-1 (Prx-1) in Dox-resistance osteosarcoma cells was detected with Western blot assay. Results In this study, the inhibitory effects of Dox on the viability and proliferation of Dox-resistance osteosarcoma cells were significantly enhanced by β-Elemene. In addition, the combination of Dox and β-Elemene markedly induced the apoptosis and oxidative stress in Dox-resistance osteosarcoma cells. Moreover, combination treatment notably downregulated the expression of Prx-1 in Dox-resistance osteosarcoma cells, indicating that combination treatment inhibited the antioxidant capacity of Dox-resistance osteosarcoma cells. In vivo experiments confirmed that β-Elemene could enhance the anti-tumor effect of Dox in Saos-2/Dox xenograft model. Conclusion We found that β-Elemene could reverse Dox resistance in Dox-resistance osteosarcoma cells via inhibition of Prx-1. Therefore, combining Dox with β-Elemene might be considered as a therapeutic approach for the treatment of Dox-resistant osteosarcoma.
Collapse
Affiliation(s)
- Shaochun Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China.,Department of Orthopedics, Ezhou Central Hospital, Ezhou, Hubei, 436000, People's Republic of China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| |
Collapse
|
30
|
Sneha KR, Sreeja S, Sailaja GS. Radiopacity endowed magnetic nanocomposite with hyperthermia and in vitromineralization potential: a combinatorial therapeutic system for osteosarcoma. Biomed Mater 2021; 16. [PMID: 34061045 DOI: 10.1088/1748-605x/ac01af] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022]
Abstract
The development of clinically advanced multifaceted therapeutic materials for osteosarcoma is at the forefront of cancer research. Accordingly, this work presents the design of a multifunctional magnetic nanocomposite composed of maghemite, strontium doped hydroxyapatite and silica nanoparticles prospectively holding indispensable therapeutic features such as magnetic hyperthermia,in vitrobiomineralization, sustained drug release and intrinsic radiopacity for the treatment of osteosarcoma. The optimal composition has been identified by sequentially modulating the ratio of precursors of the magnetic nanocomposite synthesized by sol-gel technique. Structural and morphological characterization by x-ray diffraction, fourier transform infrared spectrum, Brunauer-Emmet-Teller and transmission electron microscopy analyses followed by VSM, hyperthermia and micro-CT analyses essentially assisted in the selective configuration of biofunctional properties. Results exemplify that MSHSr1 has a saturation magnetization of 47.4 emu g-1and attained hyperthermia temperature (42 °C) at a very low exposure time of 4 min. MSHSr1 is further unique with respect to its exceptional x-ray attenuation ability (contrast enhancement 154.5% in digital radiography; CT number 3100 HU), early biomimetic mineralization (in vitro) evident by the formation of spheroidal apatite layer (Ca/P ratio 1.33) harvested from FESEM-EDX analysis and controlled release of Doxorubicin, the clinically used chemotherapeutic drug: 87.7% at 120 h in tumour analogous pH (6.5) when compared to physiological pH (71.3% at 7.4). MTT assay complemented with cytoskeleton (F-actin) staining of human osteosarcoma (HOS) cells affirm biocompatibility of MSHSr1.In vitrobiomineralization authenticated by Alizarin red S and von Kossa staining has been further corroborated by semi-quantitative calcium estimation of HOS cells cultured with MSHSr1 for two weeks. The results therefore validate the multifunctionality of MSHSr1, and hence could be proposed as a combinatorial therapeutic nanocomposite for osteosarcoma treatment.
Collapse
Affiliation(s)
- K R Sneha
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kochi 682022, India
| | - S Sreeja
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kochi 682022, India
| | - G S Sailaja
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kochi 682022, India.,Inter University Centre for Nanomaterials and Devices, CUSAT, Kochi 682022, India.,Centre for Advanced Materials, CUSAT, Kochi 682022, India
| |
Collapse
|
31
|
Barani M, Mukhtar M, Rahdar A, Sargazi S, Pandey S, Kang M. Recent Advances in Nanotechnology-Based Diagnosis and Treatments of Human Osteosarcoma. BIOSENSORS 2021; 11:55. [PMID: 33672770 PMCID: PMC7924594 DOI: 10.3390/bios11020055] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022]
Abstract
Osteosarcoma (OSA) is a type of bone cancer that begins in the cells that form bones.OSA is a rare mesenchymal bone neoplasm derived from mesenchymal stem cells. Genome disorganization, chromosomal modifications, deregulation of tumor suppressor genes, and DNA repair defects are the factors most responsible for OSA development. Despite significant advances in the diagnosing and treatment of OSA, patients' overall survival has not improved within the last twenty years. Lately, advances in modern nanotechnology have spurred development in OSA management and offered several advantages to overcome the drawbacks of conventional therapies. This technology has allowed the practical design of nanoscale devices combined with numerous functional molecules, including tumor-specific ligands, antibodies, anti-cancer drugs, and imaging probes. Thanks to their small sizes, desirable drug encapsulation efficiency, and good bioavailability, functionalized nanomaterials have found wide-spread applications for combating OSA progression. This review invokes the possible utility of engineered nanomaterials in OSA diagnosis and treatment, motivating the researchers to seek new strategies for tackling the challenges associated with it.
Collapse
Affiliation(s)
- Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 76169-14111, Iran;
| | - Mahwash Mukhtar
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, 6720 Szeged, Hungary;
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-98615, Iran
| | - Saman Sargazi
- Cellular and Molecule Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran;
| | - Sadanand Pandey
- Particulate Matter Research Center, Research Institute of Industrial Science & Technology (RIST), 187-12, Geumho-ro, Gwangyang-si 57801, Korea
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea;
| | - Misook Kang
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea;
| |
Collapse
|
32
|
Desai SA, Manjappa A, Khulbe P. Drug delivery nanocarriers and recent advances ventured to improve therapeutic efficacy against osteosarcoma: an overview. J Egypt Natl Canc Inst 2021; 33:4. [PMID: 33555490 DOI: 10.1186/s43046-021-00059-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/18/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is one of the key cancers affecting the bone tissues, primarily occurred in children and adolescence. Recently, chemotherapy followed by surgery and then post-operative adjuvant chemotherapy is widely used for the treatment of OS. However, the lack of selectivity and sensitivity to tumor cells, the development of multi-drug resistance (MDR), and dangerous side effects have restricted the use of chemotherapeutics. MAIN BODY There is an unmet need for novel drug delivery strategies for effective treatment and management of OS. Advances in nanotechnology have led to momentous progress in the design of tumor-targeted drug delivery nanocarriers (NCs) as well as functionalized smart NCs to achieve targeting and to treat OS effectively. The present review summarizes the drug delivery challenges in OS, and how organic nanoparticulate approaches are useful in overcoming barriers will be explained. The present review describes the various organic nanoparticulate approaches such as conventional nanocarriers, stimuli-responsive NCs, and ligand-based active targeting strategies tested against OS. The drug conjugates prepared with copolymer and ligand having bone affinity, and advanced promising approaches such as gene therapy, gene-directed enzyme prodrug therapy, and T cell therapy tested against OS along with their reported limitations are also briefed in this review. CONCLUSION The nanoparticulate drugs, drug conjugates, and advanced therapies such as gene therapy, and T cell therapy have promising and potential application in the effective treatment of OS. However, many of the above approaches are still at the preclinical stage, and there is a long transitional period before their clinical application.
Collapse
Affiliation(s)
- Sujit Arun Desai
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Rd, Mahal, Jagatpura, Jaipur, Rajasthan, 302017, India. .,Annasaheb Dange College of D Pharmacy, Ashta, Tal: Walwa, Dist., Sangli, Maharashtra, 416301, India.
| | - Arehalli Manjappa
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist., Kolhapur, Maharashtra, 416113, India
| | - Preeti Khulbe
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Rd, Mahal, Jagatpura, Jaipur, Rajasthan, 302017, India
| |
Collapse
|
33
|
Wang Z, Liu L, Yin W, Liu Z, Shi L, Tang M. A Novel Drug Delivery System: the Encapsulation of Naringenin in Metal-Organic Frameworks into Liposomes. AAPS PharmSciTech 2021; 22:61. [PMID: 33527250 DOI: 10.1208/s12249-021-01927-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/10/2021] [Indexed: 02/08/2023] Open
Abstract
Poorly water-soluble naringenin (NAR) was selected as a model drug and loaded into the porous MOFs for the construction of NAR@ZIF-8 inclusion complex. By film dispersion method, NAR@ZIF-8 was further encapsulated into liposomes to fabricate a novel drug delivery system. Liposomes and a novel drug delivery system were established. Subsequently, the lipid-drug ratio, phospholipid-cholesterol ratio, and hydration temperature were investigated using the Box-Behnken design based the single factor experiment. The prepared liposomes system showed spherical or quasi-spherical shape, uniform particle size distribution, and complete structure. More specifically, the average particle size was 113.2 ± 1.4 nm, and zeta potential was - 7.536 ± 0.264 mV. Moreover, the drug release behaviors of NAR, NAR@ZIF-8, and NAR@ZIF-8 liposomes were explored in vitro. Compared with free NAR and NAR@ZIF-8 which exhibited a burst drug release, NAR@ZIF-8 liposomes showed a more sustained release behavior with 79.86% drug release in 72 h. In vitro cytotoxicity experiments showed that, compared with free NAR and NAR@ZIF-8, NAR@ZIF-8 liposomes exhibited higher inhibition efficiency on lung adenocarcinoma A549 cells and gastric cancer SGC-7901 cells in a concentration-dependent manner.
Collapse
|
34
|
Akman M, Belisario DC, Salaroglio IC, Kopecka J, Donadelli M, De Smaele E, Riganti C. Hypoxia, endoplasmic reticulum stress and chemoresistance: dangerous liaisons. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:28. [PMID: 33423689 PMCID: PMC7798239 DOI: 10.1186/s13046-020-01824-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Solid tumors often grow in a micro-environment characterized by < 2% O2 tension. This condition, together with the aberrant activation of specific oncogenic patwhays, increases the amount and activity of the hypoxia-inducible factor-1α (HIF-1α), a transcription factor that controls up to 200 genes involved in neoangiogenesis, metabolic rewiring, invasion and drug resistance. Hypoxia also induces endoplasmic reticulum (ER) stress, a condition that triggers cell death, if cells are irreversibly damaged, or cell survival, if the stress is mild.Hypoxia and chronic ER stress both induce chemoresistance. In this review we discuss the multiple and interconnected circuitries that link hypoxic environment, chronic ER stress and chemoresistance. We suggest that hypoxia and ER stress train and select the cells more adapted to survive in unfavorable conditions, by activating pleiotropic mechanisms including apoptosis inhibition, metabolic rewiring, anti-oxidant defences, drugs efflux. This adaptative process unequivocally expands clones that acquire resistance to chemotherapy.We believe that pharmacological inhibitors of HIF-1α and modulators of ER stress, although characterized by low specificty and anti-cancer efficacy when used as single agents, may be repurposed as chemosensitizers against hypoxic and chemorefractory tumors in the next future.
Collapse
Affiliation(s)
- Muhlis Akman
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino, Italy
| | | | | | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Enrico De Smaele
- Department of Experimental Medicine, Sapienza University of Roma, Roma, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino, Italy.
| |
Collapse
|
35
|
Cano ME, Lesur D, Bincoletto V, Gazzano E, Stella B, Riganti C, Arpicco S, Kovensky J. Synthesis of defined oligohyaluronates-decorated liposomes and interaction with lung cancer cells. Carbohydr Polym 2020; 248:116798. [PMID: 32919536 DOI: 10.1016/j.carbpol.2020.116798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/29/2022]
Abstract
In this work hyaluronic acid (HA) oligosaccharides with degree of polymerization (DP) 4, 6 and 8, obtained by enzymatic depolymerization of HA, were conjugated to a PEG-phospholipid moiety. The products (HA-DP4, HA-DP6 and HA-DP8) were used to prepare decorated liposomes. The cellular uptake of HA-DP4, HA-DP6 and HA-DP8-decorated fluorescently labelled liposomes was significantly higher (12 to 14-fold) in lung cancer cell lines with high CD44 expression than in those with low CD44 expression, suggesting a receptor-mediated entry of HA-conjugated formulations. Competition assays showed that the uptake followed this rank order: HA-DP8>HA-DP6>HA-DP4 liposomes. Moreover, they are capable of a faster interaction with CD44, followed by phagocytosis, than HA liposomes obtained from HA of higher molecular weight (4800 and 14800 Da). HA-DP4, HA-DP6 and HA-DP8-liposomes did not show cytotoxicity or inflammatory effects. Overall, we propose our new HA-DP oligosaccharides as biocompatible and effective tools for a potential drug delivery to CD44-positive cells.
Collapse
Affiliation(s)
- Maria Emilia Cano
- Laboratoire de Glycochimie, des Antimicrobiens et des Agroressources CNRS UMR 7378, Université de Picardie Jules Verne, 33 rue Saint Leu, 80039 Amiens, France
| | - David Lesur
- Laboratoire de Glycochimie, des Antimicrobiens et des Agroressources CNRS UMR 7378, Université de Picardie Jules Verne, 33 rue Saint Leu, 80039 Amiens, France
| | - Valeria Bincoletto
- Department of Drug Science and Technology, University of Torino, Via Giuria 9, 10125 Torino, Italy
| | - Elena Gazzano
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Barbara Stella
- Department of Drug Science and Technology, University of Torino, Via Giuria 9, 10125 Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Torino, Via Giuria 9, 10125 Torino, Italy.
| | - José Kovensky
- Laboratoire de Glycochimie, des Antimicrobiens et des Agroressources CNRS UMR 7378, Université de Picardie Jules Verne, 33 rue Saint Leu, 80039 Amiens, France.
| |
Collapse
|
36
|
Zhong XC, Shi MH, Liu HN, Chen JJ, Wang TT, Lin MT, Zhang ZT, Zhou Y, Lu YY, Xu WH, Gao JQ, Xu DH, Han M, Chen YD. Mitochondrial targeted doxorubicin derivatives delivered by ROS-responsive nanocarriers to breast tumor for overcoming of multidrug resistance. Pharm Dev Technol 2020; 26:21-29. [PMID: 33070673 DOI: 10.1080/10837450.2020.1832116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multidrug resistance (MDR) is a serious challenge in chemotherapy and also a major threat to breast cancer treatment. As an intracellular energy factory, mitochondria provide energy for drug efflux and are deeply involved in multidrug resistance. Mitochondrial targeted delivery of doxorubicin can overcome multidrug resistance by disrupting mitochondrial function. By incorporating a reactive oxygen species (ROS)-responsive hydrophobic group into the backbone structure of hyaluronic acid - a natural ligand for the highly expressed CD44 receptor on tumor surfaces, a novel ROS-responsive and CD44-targeting nano-carriers was constructed. In this study, mitochondria-targeted triphenylphosphine modified-doxorubicin (TPP-DOX) and amphipathic ROS-responsive hyaluronic acid derivatives (HA-PBPE) were synthesized and confirmed by 1H NMR. The nanocarriers TPP-DOX @ HA-PBPE was prepared in a regular shape and particle size of approximately 200 nm. Compared to free DOX, its antitumor activity in vitro and tumor passive targeting in vivo has been enhanced. The ROS-responsive TPP-DOX@HA-PBPE nanocarriers system provide a promising strategy for the reverse of MDR and efficient delivery of doxorubicin derivatives into drug-resistant cancer cells.
Collapse
Affiliation(s)
- Xin-Cheng Zhong
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Ming-Han Shi
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Hui-Na Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Jie-Jian Chen
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Tian-Tian Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Meng-Ting Lin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhen-Tao Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yi Zhou
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yi-Ying Lu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Wen-Hong Xu
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Dong-Hang Xu
- Department of Pharmacy, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yi-Ding Chen
- Department of Breast Surgery, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic China
| |
Collapse
|
37
|
Masetto F, Chegaev K, Gazzano E, Mullappilly N, Rolando B, Arpicco S, Fruttero R, Riganti C, Donadelli M. MRP5 nitration by NO-releasing gemcitabine encapsulated in liposomes confers sensitivity in chemoresistant pancreatic adenocarcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118824. [PMID: 32828758 DOI: 10.1016/j.bbamcr.2020.118824] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/28/2020] [Accepted: 08/10/2020] [Indexed: 12/01/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a therapy recalcitrant disease characterized by the aberrations in multiple genes that drive pathogenesis and drug chemoresistance. In this study, we synthesize a library of seven novel nitric oxide-releasing gemcitabine pro-drugs (NO-GEMs) in order to improve the effectiveness of GEM by exploiting the therapeutic effects of NO. Among these NO-GEM pro-drugs we select 5b as the most effective compound in GEM-resistant PDAC cells. After its encapsulation in liposomes for drug delivery the intracellular NO level increases and nitration associated to activity inhibition of the multidrug resistance associated protein 5 (MRP5; ABCC5) occurs. This results in GEM intracellular accumulation and enhanced apoptotic cell death in GEM-resistant PDAC cells, which express MRP5 at higher levels than GEM-sensitive cells. Our results support the development of a new anti-tumoral strategy to efficiently affect GEM-resistant PDAC cells based on the usage of NO-GEM pro-drugs.
Collapse
Affiliation(s)
- Francesca Masetto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | | | | | - Nidula Mullappilly
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Barbara Rolando
- Department of Drug Science and Technology, University of Turin, Italy
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Italy
| | - Roberta Fruttero
- Department of Drug Science and Technology, University of Turin, Italy
| | | | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy.
| |
Collapse
|
38
|
Yang L, Zhang Y, Xie J, Zhong C, He D, Wang T, Li K, Li Y, Shi D, Abagyan R, Yang L, Zhang J. Biomimetic polysaccharide-cloaked lipidic nanovesicles/microassemblies for improving the enzymatic activity and prolonging the action time for hyperuricemia treatment. NANOSCALE 2020; 12:15222-15235. [PMID: 32639489 DOI: 10.1039/d0nr02651d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The improvement and maintenance of enzymatic activities represent major challenges. However, to address these we developed novel biomimetic polysaccharide hyaluronan (Hn)-cloaked lipidic nanovesicles (BHLN) and microassemblies (BHLNM) as enzyme carriers that function by entrapping enzymes in the core or by tethering them to the inner/outer surfaces via covalent interactions. The effectiveness of these enzyme carriers was demonstrated through an evaluation of the enzymatic activity and anti-hyperuricemia bioactivity of urate oxidase (also called uricase, Uase). We showed that Uase was effectively loaded within the BHLN/BHLNM (UHLN/UHLNM) and maintained good enzymatic bioactivity through a range of effects, including isolation from the external environment due to the vesicle-carrying (shielding effect), avoidance of recognition by the reticuloendothelial system due to Hn-cloaking (long-term effect), production of beneficial conformational changes (allosteric effect) due to a favorable internal microenvironment of construction and vesicle loading, and stabilization due to the reversible conjugation of Uase or vesicle and serum albumin (deposit effect). UHLN/UHLNM had significantly increased bioavailability (∼533% and ∼331% compared to Uase) and demonstrated greatly improved efficacy, whereby the time required for UHLN/UHLNM to lower the plasma uric acid concentration to a normal level was much shorter than that for free Uase. The interactions of the therapeutic enzyme (Uase), biomimetic membrane components (Hn and phospholipid), and serum albumin were investigated with a fluorescent probe and computational simulations to help understand the superior properties of UHLN/UHLNM.
Collapse
Affiliation(s)
- Lan Yang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Yonghong Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Jiangchuan Xie
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Cailing Zhong
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Dan He
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Tingting Wang
- Biochemistry and Molecular Biology Laboratory, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 401331, China
| | - Kailing Li
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Yao Li
- Division of Infectious Disease, Chongqing Public Health Medical Center, Chongqing 400036, China
| | - Da Shi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Lin Yang
- Department of Pharmacology, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
39
|
Coencapsulation of disulfiram and doxorubicin in liposomes strongly reverses multidrug resistance in breast cancer cells. Int J Pharm 2020; 580:119191. [DOI: 10.1016/j.ijpharm.2020.119191] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 02/06/2020] [Accepted: 02/28/2020] [Indexed: 12/19/2022]
|
40
|
Melim C, Jarak I, Veiga F, Figueiras A. The potential of micelleplexes as a therapeutic strategy for osteosarcoma disease. 3 Biotech 2020; 10:147. [PMID: 32181109 PMCID: PMC7052088 DOI: 10.1007/s13205-020-2142-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/16/2020] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS) is a rare aggressive bone, presenting low patient survival rate, high metastasis and relapse occurrence, mostly due to multi-drug resistant cells. To surpass that, the use of nanomedicine for the targeted delivery of genetic material, drugs or both have been extensively researched. In this review, we address the current situation of the disorder and some gene therapy options in the nanomedicine field that have been investigated. Among them, polymeric micelles (PM) are an advantageous therapeutic alternative highly explored for OS, as they allow for the targeted transportation of poorly water-soluble drugs to cancer cells. In addition, micelleplexes are PMs with cationic properties with promising features, such as the possibility for a dual therapy, which have made them an attractive research subject. The aim of this review article is to elucidate the application of a micelleplex formulation encapsulating the underexpressed miRNA145 to achieve an active targeting to OS cells and overcome multi-drug resistance, as a new and viable therapeutic strategy.
Collapse
Affiliation(s)
- Catarina Melim
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Ivana Jarak
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Figueiras
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
41
|
Hafez HM, Hassanein H. Montelukast ameliorates doxorubicin-induced cardiotoxicity via modulation of p-glycoprotein and inhibition of ROS-mediated TNF-α/NF-κB pathways. Drug Chem Toxicol 2020; 45:548-559. [DOI: 10.1080/01480545.2020.1730885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Heba M. Hafez
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Hanaa Hassanein
- Department of Histology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
42
|
Kaur K, Carrazzone RJ, Matson JB. The Benefits of Macromolecular/Supramolecular Approaches in Hydrogen Sulfide Delivery: A Review of Polymeric and Self-Assembled Hydrogen Sulfide Donors. Antioxid Redox Signal 2020; 32:79-95. [PMID: 31691577 PMCID: PMC6918872 DOI: 10.1089/ars.2019.7864] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/22/2019] [Accepted: 10/29/2019] [Indexed: 12/24/2022]
Abstract
Significance: Cell homeostasis and redox balance are regulated in part by hydrogen sulfide (H2S), a gaseous signaling molecule known as a gasotransmitter. Given its biological roles, H2S has promising therapeutic potential, but controlled delivery of this reactive and hazardous gas is challenging due to its promiscuity, rapid diffusivity, and toxicity at high doses. Macromolecular and supramolecular drug delivery systems are vital for the effective delivery of many active pharmaceutical ingredients, and H2S stands to benefit greatly from the tunable physical, chemical, and pharmacokinetic properties of polymeric and/or self-assembled drug delivery systems. Recent Advances: Several types of H2S-releasing macro- and supramolecular materials have been developed in the past 5 years, and the field is expanding quickly. Slow-releasing polymers, polymer assemblies, polymer nano- and microparticles, and self-assembled hydrogels have enabled triggered, sustained, and/or localized H2S delivery, and many of these materials are more potent in biological assays than analogous small-molecule H2S donors. Critical Issues: H2S plays a role in a number of (patho)physiological processes, including redox balance, ion channel regulation, modulation of inducible nitric oxide synthase, angiogenesis, blood pressure regulation, and more. Chemical tools designed to (i) deliver H2S to study these processes, and (ii) exploit H2S signaling pathways for treatment of diseases require control over the timing, rate, duration, and location of release. Future Directions: Development of new material approaches for H2S delivery that enable long-term, triggered, localized, and/or targeted delivery of the gas will enable greater understanding of this vital signaling molecule and eventually expedite its clinical application.
Collapse
Affiliation(s)
- Kuljeet Kaur
- Department of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia
| | - Ryan J. Carrazzone
- Department of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia
| | - John B. Matson
- Department of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia
| |
Collapse
|
43
|
Hattinger CM, Patrizio MP, Magagnoli F, Luppi S, Serra M. An update on emerging drugs in osteosarcoma: towards tailored therapies? Expert Opin Emerg Drugs 2019; 24:153-171. [PMID: 31401903 DOI: 10.1080/14728214.2019.1654455] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: Current treatment of conventional and non-conventional high-grade osteosarcoma (HGOS) is based on the surgical removal of primary tumor and, when possible, of metastases and local reccurrence, together with systemic pre- and post-operative chemotherapy with drugs that have been used since decades. Areas covered: This review is intended to summarize the new agents and therapeutic strategies that are under clinical evaluation in HGOS, with the aim to increase the cure probability of this highly malignant bone tumor, which has not significantly improved during the last 30-40 years. The list of drugs, compounds and treatment modalities presented and discussed here has been generated by considering only those that are included in presently ongoing and recruiting clinical trials, or which have been completed in the last 2 years with reported results, on the basis of the information obtained from different and continuously updated databases. Expert opinion: Despite HGOS is a rare tumor, several clinical trials are presently evaluating different treatment strategies, which may hopefully positively impact on the outcome of patients who experience unfavorable prognosis when treated with conventional therapies.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Maria Pia Patrizio
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Federica Magagnoli
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Silvia Luppi
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| | - Massimo Serra
- Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, IRCCS Istituto Ortopedico Rizzoli , Bologna , Italy
| |
Collapse
|
44
|
Tessaro AL, Fraix A, Pedrozo da Silva AC, Gazzano E, Riganti C, Sortino S. "Three-Bullets" Loaded Mesoporous Silica Nanoparticles for Combined Photo/Chemotherapy. NANOMATERIALS 2019; 9:nano9060823. [PMID: 31159241 PMCID: PMC6631764 DOI: 10.3390/nano9060823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/14/2019] [Accepted: 05/28/2019] [Indexed: 12/21/2022]
Abstract
This contribution reports the design, preparation, photophysical and photochemical characterization, as well as a preliminary biological evaluation of mesoporous silica nanoparticles (MSNs) covalently integrating a nitric oxide (NO) photodonor (NOPD) and a singlet oxygen (1O2) photosensitizer (PS) and encapsulating the anticancer doxorubicin (DOX) in a noncovalent fashion. These MSNs bind the NOPD mainly in their inner part and the PS in their outer part in order to judiciously exploit the different diffusion radius of the cytotoxic NO and 1O2. Furthermore this silica nanoconstruct has been devised in such a way to permit the selective excitation of the NOPD and the PS with light sources of different energy in the visible window. We demonstrate that the individual photochemical performances of the photoactive components of the MSNs are not mutually affected, and remain unaltered even in the presence of DOX. As a result, the complete nanoconstruct is able to deliver NO and 1O2 under blue and green light, respectively, and to release DOX under physiological conditions. Preliminary biological results performed using A375 cancer cells show a good tolerability of the functionalized MSNs in the dark and a potentiated activity of DOX upon irradiation, due to the effect of the NO photoreleased.
Collapse
Affiliation(s)
- André Luiz Tessaro
- Laboratory of Photochemistry, Department of Drug Sciences, University of Catania, 95125 Catania, Italy.
- Department of Chemistry, Federal University of Technology, Paraná, R. Marcílio Dias, 635, Jardim Paraíso, Apucarana 86812-460, Paraná, Brazil.
| | - Aurore Fraix
- Laboratory of Photochemistry, Department of Drug Sciences, University of Catania, 95125 Catania, Italy.
| | - Ana Claudia Pedrozo da Silva
- Department of Chemistry, Universidade Estadual de Maringá, Av. Colombo, 5.790, Maringá 87.020-900, Paraná, Brazil.
| | - Elena Gazzano
- Department of Oncology, University of Torino, Via Santena 5/bis, I-10126 Torino, Italy.
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Santena 5/bis, I-10126 Torino, Italy.
| | - Salvatore Sortino
- Laboratory of Photochemistry, Department of Drug Sciences, University of Catania, 95125 Catania, Italy.
| |
Collapse
|