1
|
Zhou Y, Camisasca A, Dominguez-Gil S, Bartkowski M, Rochfort KD, Piletti M, White A, Krizsan D, O'Connor R, Quinn SJ, Iacopino D, Eustace AJ, Giordani S. Synthesis of carbon dots from spent coffee grounds: transforming waste into potential biomedical tools. NANOSCALE 2025; 17:9947-9962. [PMID: 40067158 DOI: 10.1039/d4nr05186f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Carbon dots (CDs) are small-sized, spherical nanoparticles presenting amorphous carbon cores with nanocrystalline regions of a graphitic structure. They show unique properties such as high aqueous solubility, robust chemical inertness, and non-toxicity and can be manufactured at a relatively low cost. They are also well known for outstanding fluorescence tunability and resistance to photobleaching. Together, these properties boost their potential to act as drug delivery systems (DDSs). This work presents a low-cost synthesis of CDs by upcycling spent coffee grounds (SCGs) and transforming them into value-added products. This synthetic route eliminates the use of highly toxic heavy metals, high energy-consuming reactions and long reaction times, which can improve biocompatibility while benefiting the environment. A series of physico-chemical characterisation techniques demonstrated that these SCG-derived CDs are small-sized nanoparticles with tunable fluorescence. In vitro studies with 120 h of incubation of SCG-derived CDs demonstrated their specific antiproliferative effect on the breast cancer CAL-51 cell line, accompanied by increased reactive oxygen species (ROS) production. Importantly, no impact was observed on healthy breast, kidney, and liver cells. Confocal laser scanning microscopy confirmed the intracellular accumulation of SCG-derived CDs. Furthermore, the drug efflux pumps P-glycoprotein (P-gp) and the breast cancer resistance protein (BCRP) did not impact CD accumulation in the cancer cells.
Collapse
Affiliation(s)
- Yingru Zhou
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Adalberto Camisasca
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Sofia Dominguez-Gil
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Michał Bartkowski
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Keith D Rochfort
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Martina Piletti
- Tyndall National Institute, University College Cork, Cork, Ireland
| | - Anita White
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Dorottya Krizsan
- School of Chemistry, University College Dublin, Belfield, Dublin, Ireland
| | - Robert O'Connor
- School of Physical Sciences, Dublin City University, Glasnevin, Dublin, Ireland
| | - Susan J Quinn
- School of Chemistry, University College Dublin, Belfield, Dublin, Ireland
| | - Daniela Iacopino
- Tyndall National Institute, University College Cork, Cork, Ireland
| | - Alex J Eustace
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Silvia Giordani
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
| |
Collapse
|
2
|
Zhang J, Yang Q, Zhang Y, Zhang Q, Wan Y, Yan G, Wang X. Cancer cell membrane-coated sulindac-ortho ester nanoprodrug for inhibiting COX-2 expression and chemo-photothermal synergistic antitumor therapy. Int J Pharm 2025; 674:125460. [PMID: 40112900 DOI: 10.1016/j.ijpharm.2025.125460] [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: 10/15/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
The paper reported a cancer cell membrane bio-mimetic nanodrug to inhibit the expression of COX-2 in tumor area and realize enhanced chemo-photothermal synergistic anti-tumor effect. Ortho ester bond-coupled sulindac dimer (SU-OE) was first synthesized and co-assembled with doxorubicin to obtain pH-sensitive nanodrug (SU-OE@DOX NPs). Indocyanine green (ICG)-encapsulated H22 cell membrane vesicles (HM) were then co-extruded with SU-OE@DOX NPs to give the bio-mimetic nanoparticles (HM@I/NPs). HM@I/NPs displayed excellent stability and photothermal conversion efficiency. Compared to naked nanoparticles, the cell membrane-coated nanoparticles improved H22 cell uptake through homotypic targeting and effectively reduced internalization of macrophages. In vivo imaging results demonstrated that the nanoparticles could be enriched at tumor site and could raise the temperature of the tumor area to 56.7 °C under NIR laser irradiation. The released SU from HM@I/NPs can inhibit the expression of COX-2, and finally enhanced the chemo-PTT synergistic anti-tumor effect.
Collapse
Affiliation(s)
- Jingwen Zhang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Qirong Yang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Yiming Zhang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Qide Zhang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Yingda Wan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Guoqing Yan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China.
| |
Collapse
|
3
|
Wen Z, Luo S, Liu J, Huang Y, Chen G, Cai H. Polyallylamine Hydrochloride-Modified Bovine Serum Albumin Nanoparticles Loaded with α-Solanine for Chemotherapy of Pancreatic Cancer. Int J Nanomedicine 2025; 20:4235-4255. [PMID: 40225222 PMCID: PMC11988199 DOI: 10.2147/ijn.s508936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/29/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction α-Solanine (α-Sol) shows promise for pancreatic cancer (PC) treatment by inhibiting PC cell proliferation, migration, and invasion. However, its clinical application is hindered by poor tumor targeting, significant toxicity, and undesirable pharmacokinetics. To address these issues, this study developed a nanoparticle delivery system (PBSO NPs) using bovine serum albumin as a carrier, with polyallylamine hydrochloride surface modification to enhance α-Sol delivery. Methods PBSO NPs were characterized using transmission electron microscopy, dynamic light scattering, nanoparticle size analyzers, and Fourier-transform infrared spectroscopy. Their in vitro drug release profile and cellular uptake capabilities were evaluated. Furthermore, in vitro experiments were conducted using mouse pancreatic cancer cells (Panc02) to investigate the effects of PBSO NPs on Panc02 cell viability, migration, invasion, and apoptosis. Additionally, a pancreatic cancer xenograft tumor model was established for in vivo experiments to explore the impact of PBSO NPs on tumor growth. Results This study successfully developed PBSO NPs with favorable morphology and physiological stability, capable of enhancing cellular uptake. In vitro experiments demonstrated that PBSO NPs significantly inhibited the viability, migration, and invasion of Panc02 cells while promoting apoptosis. Moreover, PBSO NPs enhanced the inhibitory effects of α-Sol on Panc02 cells. In vivo experiments further confirmed that PBSO NPs improved the therapeutic efficacy of α-Sol against PC while partially reducing its toxicity. Additionally, PBSO NPs exhibited good biocompatibility. Discussion PBSO NPs enhance the therapeutic efficacy of α-Sol against PC by inhibiting the viability, migration, and invasion of PC cells while promoting apoptosis, thereby suppressing the progression of PC. This provides a promising therapeutic strategy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Zhengde Wen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
- Wenzhou Key Laboratory of Perioperative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Shan Luo
- Wenzhou Key Laboratory of Perioperative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
- Department of Anaesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Juntao Liu
- Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Yufan Huang
- Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Gang Chen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Huajie Cai
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
4
|
Zhra M, Akhund SA, Mohammad KS. Advancements in Osteosarcoma Therapy: Overcoming Chemotherapy Resistance and Exploring Novel Pharmacological Strategies. Pharmaceuticals (Basel) 2025; 18:520. [PMID: 40283955 PMCID: PMC12030420 DOI: 10.3390/ph18040520] [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: 02/28/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Osteosarcoma is recognized as the most prevalent primary bone malignancy, primarily affecting children and adolescents. It is characterized by its aggressive behavior and high metastatic potential, which often leads to poor patient outcomes. Despite advancements in surgical techniques and chemotherapy regimens, the prognosis for patients with osteosarcoma remains unsatisfactory, with survival rates plateauing over the past few decades. A significant barrier to effective treatment is the development of chemotherapy resistance, which complicates the management of the disease and contributes to high rates of recurrence. This review article aims to provide a comprehensive overview of recent advancements in osteosarcoma therapy, particularly in overcoming chemotherapy resistance. We begin by discussing the current standard treatment modalities, including surgical resection and conventional chemotherapy agents such as methotrexate, doxorubicin, and cisplatin. While these approaches have been foundational in managing osteosarcoma, they are often limited by adverse effects and variability in efficacy among patients. To address these challenges, we explore novel pharmacological strategies that aim to enhance treatment outcomes. This includes targeted therapies focusing on specific molecular alterations in osteosarcoma cells and immunotherapeutic approaches designed to harness the body's immune system against tumors. Additionally, we review innovative drug delivery systems that aim to improve the bioavailability and efficacy of existing treatments while minimizing toxicity. The review also assesses the mechanisms underlying chemotherapy resistance, such as drug efflux mechanisms, altered metabolism, and enhanced DNA repair pathways. By synthesizing current research findings, we aim to highlight the potential of new therapeutic agents and strategies for overcoming these resistance mechanisms. Ultimately, this article seeks to inform future research directions and clinical practices, underscoring the need for continued innovation in treating osteosarcoma to improve patient outcomes and survival rates.
Collapse
Affiliation(s)
| | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.); (S.A.A.)
| |
Collapse
|
5
|
Buyukgolcigezli I, Tenekeci AK, Sahin IH. Opportunities and Challenges in Antibody-Drug Conjugates for Cancer Therapy: A New Era for Cancer Treatment. Cancers (Basel) 2025; 17:958. [PMID: 40149295 PMCID: PMC11939980 DOI: 10.3390/cancers17060958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
The antibody, linker, and payload moieties all play a significant role in giving the ADC its unique therapeutic potential. The antibody subclass employed in ADCs is determined based on relative individual receptor affinities and pharmacokinetics. Meanwhile, the linker used in an ADC can either be cleavable or non-cleavable. ADC therapy comprises antibody-dependent mechanisms in addition to the direct cytotoxic effects of the payload. These include antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis, as well as the "bystander effect", which refers to the diffusion of a portion of the cytotoxic molecules out of the target cell, exerting its cytotoxic effect on the adjacent cells. Target antigens of ADCs are expected to be expressed on the membranes of the cancer cells facing the external matrix, although new approaches utilize antigens regarding tumor-associated cells, the tumor microenvironment, or the tumor vasculature. These target antigens of ADCs not only determine the efficacy of these agents but also impact the off-targets and related adverse effects. The majority of ADC-related toxicities are associated with off-targets. The proposed mechanisms of ADC resistance include disrupted intracellular drug trafficking, dysfunctional lysosomal processing, and the efflux of the cytotoxic molecule via ATP-binding cassette (ABC) transporters. The latter mechanism is especially prominent for multi-drug-resistant tumors. An important limitation of ADCs is the penetration of the conjugate into the tumor microenvironment and their delivery to target cancer cells. Cancerous tissues' vascular profile and the steric "binding site barrier" formed around the peripheral vessels of tumors stand as potential challenges of ADC therapy for solid tumors. As research efforts focus on reducing toxicities, overcoming resistance, and improving pharmacokinetics, ADC options for cancer therapy are expected to continue to diversify, including standalone approaches and combination therapies.
Collapse
Affiliation(s)
| | - Ates Kutay Tenekeci
- Department of Biochemistry, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey;
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ibrahim Halil Sahin
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
6
|
Yang Y, Ye T, Shang F, Chen D, Wang K, He S. Combined Albumin Polyester Nanocarriers with Docetaxel for Effective Against Lung Cancer in Mice Model. Int J Nanomedicine 2025; 20:2103-2118. [PMID: 39990292 PMCID: PMC11844320 DOI: 10.2147/ijn.s487344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Lung cancer, a deadly malignancy, often employs Docetaxel (DTX) as a chemotherapy option. However, DTX non-selective distribution limits its therapeutic effectiveness due to adverse side effects. This study aims to develop novel folate-targeted albumin polyester nanocarriers (FA-DTX-APs) encapsulating DTX for precise delivery, enhancing lung cancer treatment efficacy. Methods FA-DTX-APs were meticulously crafted utilizing the thin-film dispersion technique and subsequently evaluated for their physicochemical characteristics, encapsulation efficiency, and drug release profiles. To assess their biological properties, anti-tumor efficacy, and biosafety in the context of lung cancer, a comprehensive series of hemolysis assays, cellular studies, and animal experiments were conducted. Results FA-DTX-APs exhibit nanovesicle properties with a size of (223.65 ± 6.83) nm, a potential of (26.76 ± 3.15) mV, and encapsulate DTX with high efficiency (96.19 ± 3.27%) and loading capacity (9.75 ± 0.38%). FA-DTX-APs enable tumor-targeted drug delivery and slow release of the drug over a long period of time, with faster release in acidic environments. By efficiently targeting and entering lung cancer cells, FA-DTX-APs effectively hinder cancer growth (P < 0.05), demonstrating superior anti-tumor effects (P < 0.05), biocompatibility and enhanced biological safety (P < 0.05). Conclusion This study introduces FA-DTX-APs, an innovative nanocarrier characterized by exceptional biocompatibility and safety. It successfully targets lung cancer cells to deliver DTX in a sustained, slow-release manner, ensuring prolonged tumor-killing effects. As such, FA-DTX-APs hold immense promise as a novel nanoagent for lung cancer therapy.
Collapse
Affiliation(s)
- Yixiao Yang
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, People’s Republic of China
| | - Tao Ye
- Department of Oncology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, 201100, People’s Republic of China
| | - Fusheng Shang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, People’s Republic of China
| | - Dagui Chen
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, People’s Republic of China
| | - Kai Wang
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, People’s Republic of China
| | - Shengli He
- Department of Hepatobiliary-Pancreatic and Integrative Oncology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 200240, People’s Republic of China
| |
Collapse
|
7
|
Kontoghiorghes GJ. New Insights into Aspirin's Anticancer Activity: The Predominant Role of Its Iron-Chelating Antioxidant Metabolites. Antioxidants (Basel) 2024; 14:29. [PMID: 39857363 PMCID: PMC11763074 DOI: 10.3390/antiox14010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Epidemiological studies have suggested that following long-term, low-dose daily aspirin (LTLDA) administration for more than 5 years at 75-100 mg/day, 20-30% of patients (50-80 years old) had a lower risk of developing colorectal cancer (CRC) and about the same proportion in developing iron deficiency anemia (IDA). In cases of IDA, an increase in iron excretion is suspected, which is caused by aspirin chelating metabolites (ACMs): salicylic acid, salicyluric acid, 2,5-dihydroxybenzoic acid, and 2,3-dihydroxybenzoic acid. The ACMs constitute 70% of the administered aspirin dose and have much longer half-lives than aspirin in blood and tissues. The mechanisms of cancer risk reduction in LTLDA users is likely due to the ACM's targeting of iron involved in free radical damage, iron-containing toxins, iron proteins, and associated metabolic pathways such as ferroptosis. The ACMs from non-absorbed aspirin (about 30%) may also mitigate the toxicity of heme and nitroso-heme and other iron toxins from food, which are responsible for the cause of colorectal cancer. The mode of action of aspirin as a chelating antioxidant pro-drug of the ACMs, with continuous presence in LTLDA users, increases the prospect for prophylaxis in cancer and other diseases. It is suggested that the anticancer effects of aspirin depend primarily on the iron-chelating antioxidant activity of the ACMs. The role of aspirin in cancer and other diseases is incomplete without considering its rapid biotransformation and the longer half-life of the ACMs.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
8
|
Weissberger D, Stenzel MH, Hunter L. Precious Cargo: The Role of Polymeric Nanoparticles in the Delivery of Covalent Drugs. Molecules 2024; 29:4949. [PMID: 39459317 PMCID: PMC11510600 DOI: 10.3390/molecules29204949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Covalent drugs can offer significant advantages over non-covalent drugs in terms of pharmacodynamics (i.e., target-binding properties). However, the development of covalent drugs is sometimes hampered by pharmacokinetic limitations (e.g., low bioavailability, rapid metabolism and toxicity due to off-target binding). Polymeric nanoparticles offer a potential solution to these limitations. Delivering covalent drugs via polymeric nanoparticles provides myriad benefits in terms of drug solubility, permeability, lifetime, selectivity, controlled release and the opportunity for synergistic administration alongside other drugs. In this short review, we examine each of these benefits in turn, illustrated through multiple case studies.
Collapse
Affiliation(s)
| | - Martina H. Stenzel
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Luke Hunter
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| |
Collapse
|
9
|
Chen H, Xu R, Xu E, Chen Y, Niu C, Chen Y. Construction and performance evaluation of polyguluronic acid polysaccharides-based drug delivery systems. Colloids Surf B Biointerfaces 2024; 242:114083. [PMID: 39029246 DOI: 10.1016/j.colsurfb.2024.114083] [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: 05/06/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/21/2024]
Abstract
Polysaccharides have garnered significant attention as potential nanoparticle carriers for targeted tumor therapy due to their excellent biodegradability and biocompatibility. Polyguluronic acid (PG) is a homogeneous acidic polysaccharide fragment derived from alginate, which is found in brown algae, possesses excellent bioactivities, unique properties. This study explored the immunomodulatory activity of PG and developed PG-based nanogels through modified disulfide bonds and Ca2+ dual crosslinking. We characterized their structure, assessed their drug-loading and release properties, and ultimately validated both the safety of the nanocarrier and the in vitro anti-tumor efficacy of the encapsulated drug. Results indicated that PG significantly enhanced the proliferative activity and phagocytosis of RAW264.7 cells while promoting reactive oxygen species (ROS) production and cytokine secretion. The study identified TLR4 as the primary receptor for PG recognition in RAW264.7 cells. Furthermore, PG-based drug-carrying nanogels were prepared, exhibiting uniform sizes of about 184 nm and demonstrating exceptional encapsulation efficiency (82.15 ± 0.82 %) and drug loading capacity (8.12 ± 0.08 %). In vitro release experiments showed that these nanogels could responsively release drugs under conditions of high glutathione (GSH) reduction, facilitating drug accumulation at tumor sites and enhancing therapeutic efficacy. This research not only expands the application of PG in drug delivery systems but also provides valuable insights into leveraging natural immunomodulatory polysaccharides as carriers for targeted drug delivery.
Collapse
Affiliation(s)
- Huilin Chen
- College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, People's Republic of China
| | - Ran Xu
- College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, People's Republic of China
| | - Enyu Xu
- College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, People's Republic of China
| | - Yan Chen
- College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, People's Republic of China
| | - Chunyu Niu
- College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, People's Republic of China.
| | - Yin Chen
- College of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, People's Republic of China.
| |
Collapse
|
10
|
Romagnoli A, Rexha J, Perta N, Di Cristofano S, Borgognoni N, Venturini G, Pignotti F, Raimondo D, Borsello T, Di Marino D. Peptidomimetics design and characterization: Bridging experimental and computer-based approaches. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 212:279-327. [PMID: 40122649 DOI: 10.1016/bs.pmbts.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Peptidomimetics, designed to mimic peptide biological activity with more drug-like properties, are increasingly pivotal in medicinal chemistry. They offer enhanced systemic delivery, cell penetration, target specificity, and protection against peptidases when compared to their native peptide counterparts. Already utilized in treating diverse diseases like neurodegenerative disorders, cancer and infectious diseases, their future in medicine seems bright, with many peptidomimetics in clinical trials or development stages. Peptidomimetics are well-suited for addressing disturbed protein-protein interactions (PPIs), which often underlie various pathologies. Structural biology and computational methods like molecular dynamics simulations facilitate rational design, whereas machine learning algorithms accelerate protein structure prediction, enabling efficient drug development. Experimental validation via various spectroscopic, biophysical, and biochemical assays confirms computational predictions and guides further optimization. Peptidomimetics, with their tailored constrained structures, represent a frontier in drug design focused on targeting PPIs. In this overview, we present a comprehensive landscape of peptidomimetics, encompassing perspectives on involvement in pathologies, chemical strategies, and methodologies for their characterization, spanning in silico, in vitro and in cell approaches. With increasing interest from pharmaceutical sectors, peptidomimetics hold promise for revolutionizing therapeutic approaches, marking a new era of precision drug discovery.
Collapse
Affiliation(s)
- Alice Romagnoli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy; New York-Marche Structural Biology Centre (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy; Neuronal Death and Neuroprotection Unit, Department of Neuroscience, Mario Negri Institute for Pharmacological Research-IRCCS, Milan, Italy.
| | - Jesmina Rexha
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy; New York-Marche Structural Biology Centre (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy; Neuronal Death and Neuroprotection Unit, Department of Neuroscience, Mario Negri Institute for Pharmacological Research-IRCCS, Milan, Italy
| | - Nunzio Perta
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy; New York-Marche Structural Biology Centre (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy; Neuronal Death and Neuroprotection Unit, Department of Neuroscience, Mario Negri Institute for Pharmacological Research-IRCCS, Milan, Italy
| | | | - Noemi Borgognoni
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy; New York-Marche Structural Biology Centre (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy; Neuronal Death and Neuroprotection Unit, Department of Neuroscience, Mario Negri Institute for Pharmacological Research-IRCCS, Milan, Italy
| | - Gloria Venturini
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy; New York-Marche Structural Biology Centre (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy
| | - Francesco Pignotti
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy; New York-Marche Structural Biology Centre (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy
| | - Domenico Raimondo
- Department of Molecular Medicine, Spienza University of Rome, Rome, Italy; National Biodiversity Future Center (NBFC), Rome, Italy
| | - Tiziana Borsello
- Neuronal Death and Neuroprotection Unit, Department of Neuroscience, Mario Negri Institute for Pharmacological Research-IRCCS, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Daniele Di Marino
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy; New York-Marche Structural Biology Centre (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy; Neuronal Death and Neuroprotection Unit, Department of Neuroscience, Mario Negri Institute for Pharmacological Research-IRCCS, Milan, Italy
| |
Collapse
|
11
|
Liu Y, Zhang D, Zhang Z, Liang X, Yang X, Ding N, Nie Y, Li C. Multifunctional nanoparticles inhibit tumor and tumor-associated macrophages for triple-negative breast cancer therapy. J Colloid Interface Sci 2024; 657:598-610. [PMID: 38071809 DOI: 10.1016/j.jcis.2023.11.156] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 01/02/2024]
Abstract
HYPOTHESIS Tumor-associated macrophages (TAM) are the mainstay of immunosuppressive cells in the tumor microenvironment, and elimination of M2-type macrophages (M2-TAM) is considered as a potential immunotherapy. However, the interaction of breast cancer cells with macrophages hinders the effectiveness of immunotherapy. In order to improve the efficacy of triple-negative breast cancer (TNBC) therapy, strategies that simultaneously target the elimination of M2-TAM and breast cancer cells may be able to achieve a better therapy. EXPERIMENTS LyP-SA/AgNP@Dox multifunctional nanoparticles were synthesized by electrostatic adsorption. They were characterized by particle size, potential and spectroscopy. And the efficacy of multifunctional nanoparticles was evaluated in 4 T1 cell lines and M2 macrophages, including their cell uptake intracellular reactive oxygen species (ROS) production and the therapeutic effect. Furthermore, based on the orthotopic xenotransplantation model of triple negative breast cancer, the biological distribution, fluorescence imaging, biosafety evaluation and combined efficacy evaluation of the nanoplatform were performed. FINDINGS We have successfully prepared LyP-SA/AgNP@Dox and characterized. Administering the nanosystem to 4 T1 tumor cells or M2 macrophages in culture induced accumulation of reactive oxygen species, destruction of mitochondria and apoptosis, and inhibited replication and transcription. Animal experiments demonstrated the nanoparticle had favorable targeting and antitumor activity. Our nanosystem may be useful for simultaneously inhibiting tumor and tumor-associated macrophages in breast cancer and, potentially, other malignancies.
Collapse
Affiliation(s)
- Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Dan Zhang
- Department of Pharmacy of Traditional Chinese Medicine, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Nianhui Ding
- Department of Pharmacology Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
12
|
Zhou XQ, Li YP, Dang SS. Precision targeting in hepatocellular carcinoma: Exploring ligand-receptor mediated nanotherapy. World J Hepatol 2024; 16:164-176. [PMID: 38495282 PMCID: PMC10941735 DOI: 10.4254/wjh.v16.i2.164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 02/27/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and poses a major challenge to global health due to its high morbidity and mortality. Conventional chemotherapy is usually targeted to patients with intermediate to advanced stages, but it is often ineffective and suffers from problems such as multidrug resistance, rapid drug clearance, nonspecific targeting, high side effects, and low drug accumulation in tumor cells. In response to these limitations, recent advances in nanoparticle-mediated targeted drug delivery technologies have emerged as breakthrough approaches for the treatment of HCC. This review focuses on recent advances in nanoparticle-based targeted drug delivery systems, with special attention to various receptors overexpressed on HCC cells. These receptors are key to enhancing the specificity and efficacy of nanoparticle delivery and represent a new paradigm for actively targeting and combating HCC. We comprehensively summarize the current understanding of these receptors, their role in nanoparticle targeting, and the impact of such targeted therapies on HCC. By gaining a deeper understanding of the receptor-mediated mechanisms of these innovative therapies, more effective and precise treatment of HCC can be achieved.
Collapse
Affiliation(s)
- Xia-Qing Zhou
- Department of Infectious Diseases, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ya-Ping Li
- Department of Infectious Diseases, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Shuang-Suo Dang
- Department of Infectious Diseases, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| |
Collapse
|
13
|
Wilczak M, Surman M, Przybyło M. The Role of Intracellular and Extracellular Vesicles in the Development of Therapy Resistance in Cancer. Curr Pharm Des 2024; 30:2765-2784. [PMID: 39113303 DOI: 10.2174/0113816128326325240723051625] [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: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 10/22/2024]
Abstract
Cancer is the second leading cause of global mortality and claims approximately 10 million lives annually. Despite advances in treatments such as surgery, chemotherapy, and immunotherapy, resistance to these methods has emerged. Multidrug resistance (MDR), where cancer cells resist diverse treatments, undermines therapy effectiveness, escalating mortality rates. MDR mechanisms include, among others, drug inactivation, reduced drug uptake, enhanced DNA repair, and activation of survival pathways such as autophagy. Moreover, MDR mechanisms can confer resistance to other therapies like radiotherapy. Understanding these mechanisms is crucial for improving treatment efficacy and identifying new therapeutic targets. Extracellular vesicles (EVs) have gathered attention for their role in cancer progression, including MDR development through protein transfer and genetic reprogramming. Autophagy, a process balancing cellular resources, also influences MDR. The intersection of EVs and autophagy further complicates the understanding of MDR. Both extracellular (exosomes, microvesicles) and intracellular (autophagic) vesicles contribute to therapy resistance by regulating the tumor microenvironment, facilitating cell communication, and modulating drug processing. While much is known about these pathways, there is still a need to explore their potential for predicting treatment responses and understanding tumor heterogeneity.
Collapse
Affiliation(s)
- Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Magdalena Surman
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| |
Collapse
|
14
|
Mironov VF, Dimukhametov MN, Nemtarev AV, Pashirova TN, Tsepaeva OV, Voloshina AD, Vyshtakalyuk AB, Litvinov IA, Lyubina AP, Sapunova AS, Abramova DF, Zobov VV. Novel Mitochondria-Targeted Amphiphilic Aminophosphonium Salts and Lipids Nanoparticles: Synthesis, Antitumor Activity and Toxicity. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2840. [PMID: 37947686 PMCID: PMC10649961 DOI: 10.3390/nano13212840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/17/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
The creation of mitochondria-targeted vector systems is a new tool for the treatment of socially significant diseases. Phosphonium groups provide targeted delivery of drugs through biological barriers to organelles. For this purpose, a new class of alkyl(diethylAmino)(Phenyl) Phosphonium halides (APPs) containing one, two, or three diethylamino groups was obtained by the reaction of alkyl iodides (bromides) with (diethylamino)(phenyl)phosphines under mild conditions (20 °C) and high yields (93-98%). The structure of APP was established by NMR and XRD. A high in vitro cytotoxicity of APPs against M-HeLa, HuTu 80, PC3, DU-145, PANC-1, and MCF-7 lines was found. The selectivity index is in the range of 0.06-4.0 μM (SI 17-277) for the most active APPs. The effect of APPs on cancer cells is characterized by hyperproduction of ROS and depolarization of the mitochondrial membrane. APPs induce apoptosis, proceeding along the mitochondrial pathway. Incorporation of APPs into lipid systems (liposomes and solid lipid nanoparticles) improves cytotoxicity toward tumor cells and decrease toxicity against normal cell lines. The IC50s of lipid systems are lower than for the reference drug DOX, with a high SI (30-56) toward MCF-7 and DU-145. APPs exhibit high selective activity against Gram-positive bacteria S. aureus 209P and B. segeus 8035, including methicillin-resistant S. aureus (MRSA-1, MRSA-2), comparable to the activity of the fluoroquinolone antibiotic norfloxacin. A moderate in vivo toxicity in CD-1 mice was established for the lead APP.
Collapse
Affiliation(s)
- Vladimir F. Mironov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
- Alexander Butlerov Institute of Chemistry, Kazan (Volga Region) Federal University, 18 Kremlevskaya St., 420008 Kazan, Russia
| | - Mudaris N. Dimukhametov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Andrey V. Nemtarev
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
- Alexander Butlerov Institute of Chemistry, Kazan (Volga Region) Federal University, 18 Kremlevskaya St., 420008 Kazan, Russia
| | - Tatiana N. Pashirova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Olga V. Tsepaeva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Alexandra D. Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Alexandra B. Vyshtakalyuk
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Igor A. Litvinov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Anna P. Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Anastasiia S. Sapunova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Dinara F. Abramova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Vladimir V. Zobov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| |
Collapse
|
15
|
Ji P, Yang K, Xu Q, Qin G, Zhu Q, Qian Y, Yao W. Mechanisms and Application of Gas-Based Anticancer Therapies. Pharmaceuticals (Basel) 2023; 16:1394. [PMID: 37895865 PMCID: PMC10609769 DOI: 10.3390/ph16101394] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer is still one of the major factors threatening public health, with morbidity and mortality rates at the forefront of the world. Clinical drawbacks, such as high toxicity and side effects of drug therapy, and easy recurrence after surgery affect its therapeutic effect. Gas signaling molecules are essential in maintaining biological homeostasis and physiological functions as specific chemical substances for biological information transfer. In recent years, the physiological regulatory functions of gas molecules in the cancer process have been gradually revealed and have shown broad application prospects in tumor therapy. In this paper, standard gas therapies are classified and introduced. Taking H2, CO2, NO, CO, H2S, and SO2 gases as examples, the research progress and application of gas therapies in malignant tumors are mainly introduced in terms of biological characteristics, anticancer mechanisms, and treatment strategies. Finally, the problems and prospects for developing gases as anticancer drugs are outlined.
Collapse
Affiliation(s)
- Peng Ji
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Kexin Yang
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Qingqing Xu
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Guilin Qin
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Qianyu Zhu
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Ying Qian
- College of Pharmacy and Chemistry & Chemical Engineering, Jiangsu Provincial Key Laboratory of Chiral Pharmaceutical Chemicals Biologically Manufacturing, Taizhou University, Taizhou 225300, China
| | - Wenshui Yao
- Department of Anesthesiology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
16
|
Yang F, Fan Z, Zhang L, He Y, Hu R, Xiang J, Fu S, Wang G, Wang J, Tao X, Zhang P. Preparation and anti-triple-negative breast cancer cell effect of a nanoparticle for the codelivery of paclitaxel and gemcitabine. DISCOVER NANO 2023; 18:119. [PMID: 37735318 PMCID: PMC10513990 DOI: 10.1186/s11671-023-03899-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
Amphiphilic polymers (HA-ANI) were prepared by grafting hyaluronic acid (HA) and 6-(2-nitroimidazole)hexylamine (ANI) and then self-assemble in water to form nanoparticles (NPs) that could be loaded with paclitaxel (PTX) and gemcitabine (GEM) by dialysis. Infrared spectroscopy and 1H-NMR indicated the successful synthesis of HA-ANI. Three different ratios of NPs were prepared by adjusting the ratios of hydrophilic and hydrophobic materials, and the particle size decreased as the ratio of hydrophilic materials increased. When HA:ANI = 2.0:1, the nanoparticles had the smallest size distribution, good stability and near spherical shape and had high drug loading and encapsulation rates. In vitro release experiments revealed that NADPH could accelerate the drug release from NPs. Cellular uptake rate reached 86.50% at 6 h. The toxic effect of dual drug-loaded nanoparticles (P/G NPs) on MDA-MB-231 cells at 48 h was stronger than that of the free drug. The AO/EB double-staining assay revealed that a large number of late apoptotic cells appeared in the P/G NPs group, and the degree of cell damage was significantly stronger than that of the free drug group. In the cell migration assay, the 24 h-cell migration rate of the P/G NPs group was 5.99%, which was much lower than that of the free group (13.87% and 17.00%). In conclusion, MDA-MB-231 cells could effectively take up P/G NPs, while the introduction of the nano-codelivery system could significantly enhance the toxicity of the drug to MDA-MB-231 cells as well as the migration inhibition effect.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Zehui Fan
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Lixia Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yanjuan He
- Department of Pediatrics, The Fourth Hospital of Changsha, 70 Lushan Road, Changsha, 410006, Hunan, China
| | - Run Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jinkun Xiang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shiyang Fu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Guowei Wang
- Department of Spine Surgery and Department of Infection, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jianlong Wang
- Department of Spine Surgery and Department of Infection, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiaojun Tao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province and Department of Pharmacy, School of Medicine, Hunan Normal University, 371 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Pan Zhang
- Department of Spine Surgery and Department of Infection, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
17
|
Dong J, Yuan L, Hu C, Cheng X, Qin JJ. Strategies to overcome cancer multidrug resistance (MDR) through targeting P-glycoprotein (ABCB1): An updated review. Pharmacol Ther 2023; 249:108488. [PMID: 37442207 DOI: 10.1016/j.pharmthera.2023.108488] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023]
Abstract
The emergence of multidrug resistance (MDR) in malignant tumors is one of the leading threats encountered currently in many chemotherapeutic agents. The overexpression of the ATP-binding cassette (ABC) transporters is involved in MDR. P-glycoprotein (P-gp)/ABCB1 is a member of the ABC transporter family that significantly increases the efflux of various anticancer drugs from tumor cells. Therefore, targeting P-gp with small molecule inhibitors is an effective therapeutic strategy to overcome MDR. Over the past four decades, diverse compounds with P-gp inhibitory activity have been identified to sensitize drug-resistant cells, but none of them has been proven clinically useful to date. Research efforts continue to discover an effective approach for circumventing MDR. This review has provided an overview of the most recent advances (last three years) in various strategies for circumventing MDR mediated by P-gp. It may be helpful for the scientists working in the field of drug discovery to further synthesize and discover new chemical entities/therapeutic modalities with less toxicity and more efficacies to overcome MDR in cancer chemotherapy.
Collapse
Affiliation(s)
- Jinyun Dong
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| | - Li Yuan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
| | - Can Hu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
| | - Xiangdong Cheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| | - Jiang-Jiang Qin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
18
|
Sepahi S, Kiaei L, Kiaei M, Ghorani-Azam A. A systematic review of emerging technologies to enhance the treatment of ovarian cancer. Pharm Dev Technol 2023; 28:660-677. [PMID: 37417773 DOI: 10.1080/10837450.2023.2233588] [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: 03/29/2023] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
The efficacy and safety of chemotherapy are two major challenges when it comes to treating ovarian cancer. The associated undesirable side effects of chemotherapy agents jeopardize the clinical intent and the efficiency of the therapy. Multiple studies have been published describing new developments and novel strategies utilizing the latest therapeutic and drug delivery technologies to address the efficacy and safety of chemotherapeutics in ovarian cancers. We have identified five novel technologies that are available and, if used, have the potential to mitigate the above-mentioned challenges. Nanocarriers in different forms (Nano-gel, Aptamer, peptide medicated formulations, Antibody-drug conjugation, surface charge, and nanovesicle technologies) are developed and available to be employed to target the cancerous tissue. These strategies are promising to improve clinical efficacy and reduce side effects. We have systematically searched and analyzed published data, as well as the authors intent for the described technology on each publication. We narrowed to 81 key articles and extracted their data to be discussed in this review. In summary, the selected articles investigated the pharmacokinetic properties of drugs combined with nanocarriers and found significant improvement in efficacy and safety by reducing the IC50 values and drug doses. These key papers described promising novel technologies in anti-cancer therapeutic approaches to enable sustained drug release and achieve prolonged drug performance near the tumor site or target tissue.
Collapse
Affiliation(s)
- Samaneh Sepahi
- Food and Beverages Safety Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Lily Kiaei
- RockGen Therapeutics, LLC, Little Rock, AR, USA
- University of California Los Angeles, Los Angeles, CA, USA
| | - Mahmoud Kiaei
- RockGen Therapeutics, LLC, Little Rock, AR, USA
- Department of Pharmacology and Toxicology, Department of Neurology, Department of Geriatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adel Ghorani-Azam
- Department of Forensic Medicine and Toxicology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
19
|
Sun X, Zhao P, Lin J, Chen K, Shen J. Recent advances in access to overcome cancer drug resistance by nanocarrier drug delivery system. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:390-415. [PMID: 37457134 PMCID: PMC10344729 DOI: 10.20517/cdr.2023.16] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 07/18/2023]
Abstract
Cancer is currently one of the most intractable diseases causing human death. Although the prognosis of tumor patients has been improved to a certain extent through various modern treatment methods, multidrug resistance (MDR) of tumor cells is still a major problem leading to clinical treatment failure. Chemotherapy resistance refers to the resistance of tumor cells and/or tissues to a drug, usually inherent or developed during treatment. Therefore, an urgent need to research the ideal drug delivery system to overcome the shortcoming of traditional chemotherapy. The rapid development of nanotechnology has brought us new enlightenments to solve this problem. The novel nanocarrier provides a considerably effective treatment to overcome the limitations of chemotherapy or other drugs resulting from systemic side effects such as resistance, high toxicity, lack of targeting, and off-target. Herein, we introduce several tumor MDR mechanisms and discuss novel nanoparticle technology applied to surmount cancer drug resistance. Nanomaterials contain liposomes, polymer conjugates, micelles, dendrimers, carbon-based, metal nanoparticles, and nucleotides which can be used to deliver chemotherapeutic drugs, photosensitizers, and small interfering RNA (siRNA). This review aims to elucidate the advantages of nanomedicine in overcoming cancer drug resistance and discuss the latest developments.
Collapse
Affiliation(s)
- Xiangyu Sun
- Medicines and Equipment Department, Beijing Chaoyang Emergency Medical Rescuing Center, Chaoyang District, Beijing 100026, China
| | - Ping Zhao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Meg Centre, Guangzhou 510006, Guangdong, China
| | - Jierou Lin
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Meg Centre, Guangzhou 510006, Guangdong, China
| | - Kun Chen
- Beijing Chaoyang Emergency Medical Rescuing Center, Chaoyang District, Beijing 100026, China
| | - Jianliang Shen
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China
| |
Collapse
|
20
|
Mdlovu NV, Juang RS, Weng MT, Lin YS, Lin KS. Dual pH-/Thermoresponsive Shell-Cross-Linked Magnetic Mesoporous Nanospheres for Doxorubicin Delivery and In Vitro/ In Vivo Cancer Treatment. ACS APPLIED NANO MATERIALS 2023; 6:8416-8433. [DOI: 10.1021/acsanm.3c00705] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Ndumiso Vukile Mdlovu
- Department of Chemical and Materials Engineering, Chang Gung University, Guishan, Taoyuan 33302, Taiwan
- Division of Nephrology, Department of Internal Medicine, Chang Gung Memorial Hospital Linkou, Taoyuan 33305, Taiwan
| | - Ruey-Shin Juang
- Department of Chemical and Materials Engineering, Chang Gung University, Guishan, Taoyuan 33302, Taiwan
- Division of Nephrology, Department of Internal Medicine, Chang Gung Memorial Hospital Linkou, Taoyuan 33305, Taiwan
- Department of Safety, Health and Environmental Engineering, Ming Chi University of Technology, Taishan, New Taipei City 24301, Taiwan
| | - Meng-Tzu Weng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100233, Taiwan
- Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302, Taiwan
| | - You-Sheng Lin
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung−Li District, Taoyuan 32003, Taiwan
| | - Kuen-Song Lin
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung−Li District, Taoyuan 32003, Taiwan
| |
Collapse
|
21
|
Zhang S, Dong J, Pan R, Xu Z, Li M, Zang R. Structures, Properties, and Bioengineering Applications of Alginates and Hyaluronic Acid. Polymers (Basel) 2023; 15:2149. [PMID: 37177293 PMCID: PMC10181120 DOI: 10.3390/polym15092149] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
In recent years, polymeric materials have been used in a wide range of applications in a variety of fields. In particular, in the field of bioengineering, the use of natural biomaterials offers a possible new avenue for the development of products with better biocompatibility, biodegradability, and non-toxicity. This paper reviews the structural and physicochemical properties of alginate and hyaluronic acid, as well as the applications of the modified cross-linked derivatives in tissue engineering and drug delivery. This paper summarizes the application of alginate and hyaluronic acid in bone tissue engineering, wound dressings, and drug carriers. We provide some ideas on how to replace or combine alginate-based composites with hyaluronic-acid-based composites in tissue engineering and drug delivery to achieve better eco-economic value.
Collapse
Affiliation(s)
- Shuping Zhang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.D.)
| | | | | | | | | | | |
Collapse
|
22
|
Liu Z, Liu S, Zhao X, Xue C, Liu Y, Shuai Q. Photothermal-accelerated urease-powered human serum albumin nanomotor for rapid and efficient photothermal and photodynamic cancer combination therapy. Int J Biol Macromol 2023; 240:124486. [PMID: 37076068 DOI: 10.1016/j.ijbiomac.2023.124486] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Nanomotors, as a new type of micro-device, show good performance in terms of rapid transportation and deep penetration through their autonomous motion. However, their ability to efficiently break physiological barriers still remains a great challenge. Herein, we first developed a thermal-accelerated urease driven human serum albumin (HSA) nanomotor based on photothermal intervention (PTI) to achieve chemotherapy drugfree-phototherapy. The HANM@FI (HSA-AuNR@FA@Ur@ICG) is composed of a main body of biocompatible HSA, modified by gold nanorods (AuNR) and loaded with functional molecules of folic acid (FA) and indocyanine green (ICG). It promotes its own motion by breaking down urea to produce carbon dioxide and ammonia. In particular, the nanomotor is conveniently operated via near-infrared combined photothermal therapy (PTT)/ photodynamic therapy (PDT) to achieve an accelerated De value from 0.73 μm2s-1 to 1.01μm2s-1, and ideal tumor ablation at the same time. In contrast to customary urease-driven nanodrug-stacked engine, this HANM@FI has both targeting and imaging-guided capabilities, and finally achieves superior anti-tumor effects without chemotherapy drugs, through a "two-in-one" (motor mobility plus unique phototherapy in chemotherapy-drugfree phototherapy) strategy. This PTI effect with urease-driven nanomotors may offer further possibilities for future clinical applications of nanomedicines by enabling deep penetration and a subsequent chemotherapy-drugfree combination therapy strategy.
Collapse
Affiliation(s)
- Zhicheng Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Shupeng Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaoyu Zhao
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Chenglong Xue
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yu Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Qi Shuai
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
23
|
Grzes PA, Sawicka A, Niemirowicz-Laskowska K, Wielgat P, Sawicka D, Car H, Jastrzebska I. Metal-promoted synthesis of steroidal ethynyl selenides having anticancer activity. J Steroid Biochem Mol Biol 2023; 227:106232. [PMID: 36476636 DOI: 10.1016/j.jsbmb.2022.106232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
In this study, we have described simple and efficient methodology for the metal-promoted (Cu2I2) preparation of steroidal ethynyl selenides. The compounds were characterized using 1H, 13C and 77Se NMR, FT IR spectroscopy, and MS analysis. A proposed mechanism of the metal-promoted reaction involves the formation of a σ-bound copper acetylide. Due to the fact that organoselenium-based compounds possess a pleiotropic properties and associated with their promising biological activities, in the next step of the study biocompatibility and anticancer activity of the synthesized compounds was evaluated. Steroidal selenides were tested in vitro against estrogen-depend breast cancer cells MCF-7 using spectrophotometric, fluorometric and luminometric methods. Designed selenides showed high hemocompatibility, lack of toxicity against cardiomyocytes cell and great anti-cancer activity in vitro against estrogen-depend breast cancer cells upon 24 h of treatment. We revealed that selenides decrease the viability and proliferation ability of MCF-7 cells by induction of cell apoptosis. It has been noted that the overproduction of reactive oxygen species (ROS) and associated with its activation of Caspase 3/7 are a major mechanism that is responsible of selenides-caused cell death. These data indicate that organoselenium based compounds have great antineoplastic potential and might be developed as novel class of agents dedicated to the breast-cancer therapies.
Collapse
Affiliation(s)
- Pawel A Grzes
- Faculty of Chemistry, University of Białystok, ul. Ciołkowskiego 1 K, 15-245 Białystok, Poland
| | - Agata Sawicka
- Faculty of Chemistry, University of Białystok, ul. Ciołkowskiego 1 K, 15-245 Białystok, Poland
| | | | - Przemysław Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Bialystok, Poland
| | - Diana Sawicka
- Department of Experimental Pharmacology, Medical University of Bialystok, ul. Szpitalna 37, 15-295 Białystok, Poland
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, ul. Szpitalna 37, 15-295 Białystok, Poland; Department of Clinical Pharmacology, Medical University of Bialystok, Bialystok, Poland
| | - Izabella Jastrzebska
- Faculty of Chemistry, University of Białystok, ul. Ciołkowskiego 1 K, 15-245 Białystok, Poland.
| |
Collapse
|
24
|
Guo NK, She H, Tan L, Zhou YQ, Tang CQ, Peng XY, Ma CH, Li T, Liu LM. Nano Parthenolide Improves Intestinal Barrier Function of Sepsis by Inhibiting Apoptosis and ROS via 5-HTR2A. Int J Nanomedicine 2023; 18:693-709. [PMID: 36816330 PMCID: PMC9930579 DOI: 10.2147/ijn.s394544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
Background Intestinal barrier dysfunction is an important complication of sepsis, while the treatment is limited. Recently, parthenolide (PTL) has attracted much attention as a strategy of sepsis, but whether nano parthenolide (Nano PTL) is therapeutic in sepsis-induced intestinal barrier dysfunction is obscured. Methods In this study, cecal ligation and puncture (CLP)-induced sepsis rats and lipopolysaccharide (LPS)-stimulated intestinal epithelial cells (IECs) were used to investigate the effect of PTL on intestinal barrier dysfunction. Meanwhile, we synthesized Nano PTL and compared the protective effect of Nano PTL with ordinary PTL on intestinal barrier function in septic rats and IECs. Network pharmacology and serotonin 2A (5-HTR2A) inhibitor were used to explore the mechanism of PTL on the intestinal barrier function of sepsis. Results The encapsulation rate of Nano PTL was 95±1.5%, the drug loading rate was 11±0.5%, and the average uptake rate of intestinal epithelial cells was 94%. Ordinary PTL and Nano PTL improved the survival rate and survival time of septic rats, reduced the mean arterial pressure and the serum level of inflammatory cytokines, and protected the liver and kidney functions in vivo, and increased the value of transmembrane resistance (TEER) reduced the reactive oxygen species (ROS) and apoptosis in IECs in vitro through 5-HTR2A. Nano PTL had better effect than ordinary PTL. Conclusion Ordinary PTL and Nano PTL can protect the intestinal barrier function of septic rats by inhibiting apoptosis and ROS through up-regulating 5-HTR2A, Nano PTL is better than ordinary PTL.
Collapse
Affiliation(s)
- Ning-Ke Guo
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China,The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Han She
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Lei Tan
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Yuan-Qun Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Chun-Qiong Tang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Xiao-Yong Peng
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Chun-Hua Ma
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China,Correspondence: Tao Li; Liang-Ming Liu, Email ;
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
25
|
Condello M, Vona R, Meschini S. Prunus spinosa Extract Sensitized HCT116 Spheroids to 5-Fluorouracil Toxicity, Inhibiting Autophagy. Int J Mol Sci 2022; 23:ijms232416098. [PMID: 36555736 PMCID: PMC9785163 DOI: 10.3390/ijms232416098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Autophagy is a lysosomal degradation and recycling process involved in tumor progression and drug resistance. The aim of this work was to inhibit autophagy and increase apoptosis in a 3D model of human colorectal cancer by combined treatment with our patented natural product Prunus spinosa + nutraceutical activator complex (PsT + NAC®) and 5-fluorouracil (5-FU). By means of cytotoxic evaluation (MTT assay), cytofluorimetric analysis, light and fluorescence microscopy investigation and Western blotting evaluation of the molecular pathway PI3/AKT/mTOR, Caspase-9, Caspase-3, Beclin1, p62 and LC3, we demonstrated that the combination PsT + NAC® and 5-FU significantly reduces autophagy by increasing the apoptotic phenomenon. These results demonstrate the importance of using non-toxic natural compounds to improve the therapeutic efficacy and reduce the side effects induced by conventional drugs in human colon cancer.
Collapse
Affiliation(s)
- Maria Condello
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy
- Correspondence: (M.C.); (S.M.)
| | - Rosa Vona
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy
| | - Stefania Meschini
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy
- Correspondence: (M.C.); (S.M.)
| |
Collapse
|
26
|
Gomari MM, Abkhiz S, Pour TG, Lotfi E, Rostami N, Monfared FN, Ghobari B, Mosavi M, Alipour B, Dokholyan NV. Peptidomimetics in cancer targeting. Mol Med 2022; 28:146. [PMID: 36476230 PMCID: PMC9730693 DOI: 10.1186/s10020-022-00577-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
The low efficiency of treatment strategies is one of the main obstacles to developing cancer inhibitors. Up to now, various classes of therapeutics have been developed to inhibit cancer progression. Peptides due to their small size and easy production compared to proteins are highly regarded in designing cancer vaccines and oncogenic pathway inhibitors. Although peptides seem to be a suitable therapeutic option, their short lifespan, instability, and low binding affinity for their target have not been widely applicable against malignant tumors. Given the peptides' disadvantages, a new class of agents called peptidomimetic has been introduced. With advances in physical chemistry and biochemistry, as well as increased knowledge about biomolecule structures, it is now possible to chemically modify peptides to develop efficient peptidomimetics. In recent years, numerous studies have been performed to the evaluation of the effectiveness of peptidomimetics in inhibiting metastasis, angiogenesis, and cancerous cell growth. Here, we offer a comprehensive review of designed peptidomimetics to diagnose and treat cancer.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shadi Abkhiz
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Taha Ghantab Pour
- grid.411746.10000 0004 4911 7066Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Lotfi
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Rostami
- grid.411425.70000 0004 0417 7516Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, Iran
| | - Fatemeh Nafe Monfared
- grid.411705.60000 0001 0166 0922Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Ghobari
- grid.412831.d0000 0001 1172 3536Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mona Mosavi
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behruz Alipour
- grid.411705.60000 0001 0166 0922Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikolay V. Dokholyan
- grid.240473.60000 0004 0543 9901Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA ,grid.240473.60000 0004 0543 9901Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA USA
| |
Collapse
|
27
|
New Iron Metabolic Pathways and Chelation Targeting Strategies Affecting the Treatment of All Types and Stages of Cancer. Int J Mol Sci 2022; 23:ijms232213990. [PMID: 36430469 PMCID: PMC9696688 DOI: 10.3390/ijms232213990] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
There is new and increasing evidence from in vitro, in vivo and clinical studies implicating the pivotal role of iron and associated metabolic pathways in the initiation, progression and development of cancer and in cancer metastasis. New metabolic and toxicity mechanisms and pathways, as well as genomic, transcription and other factors, have been linked to cancer and many are related to iron. Accordingly, a number of new targets for iron chelators have been identified and characterized in new anticancer strategies, in addition to the classical restriction of/reduction in iron supply, the inhibition of transferrin iron delivery, the inhibition of ribonucleotide reductase in DNA synthesis and high antioxidant potential. The new targets include the removal of excess iron from iron-laden macrophages, which affects anticancer activity; the modulation of ferroptosis; ferritin iron removal and the control of hyperferritinemia; the inhibition of hypoxia related to the role of hypoxia-inducible factor (HIF); modulation of the function of new molecular species such as STEAP4 metalloreductase and the metastasis suppressor N-MYC downstream-regulated gene-1 (NDRG1); modulation of the metabolic pathways of oxidative stress damage affecting mitochondrial function, etc. Many of these new, but also previously known associated iron metabolic pathways appear to affect all stages of cancer, as well as metastasis and drug resistance. Iron-chelating drugs and especially deferiprone (L1), has been shown in many recent studies to fulfill the role of multi-target anticancer drug linked to the above and also other iron targets, and has been proposed for phase II trials in cancer patients. In contrast, lipophilic chelators and their iron complexes are proposed for the induction of ferroptosis in some refractory or recurring tumors in drug resistance and metastasis where effective treatments are absent. There is a need to readdress cancer therapy and include therapeutic strategies targeting multifactorial processes, including the application of multi-targeting drugs involving iron chelators and iron-chelator complexes. New therapeutic protocols including drug combinations with L1 and other chelating drugs could increase anticancer activity, decrease drug resistance and metastasis, improve treatments, reduce toxicity and increase overall survival in cancer patients.
Collapse
|
28
|
Electrochemotherapy: An Alternative Strategy for Improving Therapy in Drug-Resistant SOLID Tumors. Cancers (Basel) 2022; 14:cancers14174341. [PMID: 36077875 PMCID: PMC9454613 DOI: 10.3390/cancers14174341] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Chemotherapy is becoming an increasingly difficult antitumor therapy to practice due to the multiple mechanisms of drug resistance. To overcome the problem, it is possible to use alternative techniques, such as electrochemotherapy, which involves the simultaneous administration of the electrical pulse (electroporation) and the treatment with the drug in order to improve the effectiveness of the drug against the tumor. Electroporation has improved the efficacy of some chemotherapeutic agents, such bleomycin, cisplatin, mitomycin C, and 5-fluorouracil. The results of in vitro, veterinary, and clinical oncology studies are promising on various cancers, such as metastatic melanoma. The purpose of this review is to give an update on the state of the art of electrochemotherapy against the main solid tumors in the preclinical, clinical, and veterinary field. Abstract Electrochemotherapy (ECT) is one of the innovative strategies to overcome the multi drug resistance (MDR) that often occurs in cancer. Resistance to anticancer drugs results from a variety of factors, such as genetic or epigenetic changes, an up-regulated outflow of drugs, and various cellular and molecular mechanisms. This technology combines the administration of chemotherapy with the application of electrical pulses, with waveforms capable of increasing drug uptake in a non-toxic and well tolerated mechanical system. ECT is used as a first-line adjuvant therapy in veterinary oncology, where it improves the efficacy of many chemotherapeutic agents by increasing their uptake into cancer cells. The chemotherapeutic agents that have been enhanced by this technique are bleomycin, cisplatin, mitomycin C, and 5-fluorouracil. After their use, a better localized control of the neoplasm has been observed. In humans, the use of ECT was initially limited to local palliative therapy for cutaneous metastases of melanoma, but phase I/II studies are currently ongoing for several histotypes of cancer, with promising results. In this review, we described the preclinical and clinical use of ECT on drug-resistant solid tumors, such as head and neck squamous cell carcinoma, breast cancer, gynecological cancer and, finally, colorectal cancer.
Collapse
|
29
|
Curcio M, Vittorio O, Bell JL, Iemma F, Nicoletta FP, Cirillo G. Hyaluronic Acid within Self-Assembling Nanoparticles: Endless Possibilities for Targeted Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12162851. [PMID: 36014715 PMCID: PMC9413373 DOI: 10.3390/nano12162851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 05/27/2023]
Abstract
Self-assembling nanoparticles (SANPs) based on hyaluronic acid (HA) represent unique tools in cancer therapy because they combine the HA targeting activity towards cancer cells with the advantageous features of the self-assembling nanosystems, i.e., chemical versatility and ease of preparation and scalability. This review describes the key outcomes arising from the combination of HA and SANPs, focusing on nanomaterials where HA and/or HA-derivatives are inserted within the self-assembling nanostructure. We elucidate the different HA derivatization strategies proposed for this scope, as well as the preparation methods used for the fabrication of the delivery device. After showing the biological results in the employed in vivo and in vitro models, we discussed the pros and cons of each nanosystem, opening a discussion on which approach represents the most promising strategy for further investigation and effective therapeutic protocol development.
Collapse
Affiliation(s)
- Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Orazio Vittorio
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Jessica Lilian Bell
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Francesca Iemma
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| |
Collapse
|