Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.602
Filter
1.
Int J Nanomedicine ; 19: 6399-6425, 2024.
Article in English | MEDLINE | ID: mdl-38952676

ABSTRACT

Myocardial infarction, usually caused by the rupture of atherosclerotic plaque, leads to irreversible ischemic cardiomyocyte death within hours followed by impaired cardiac performance or even heart failure. Current interventional reperfusion strategies for myocardial infarction still face high mortality with the development of heart failure. Nanomaterial-based therapy has made great progress in reducing infarct size and promoting cardiac repair after MI, although most studies are preclinical trials. This review focuses primarily on recent progress (2016-now) in the development of various nanomedicines in the treatment of myocardial infarction. We summarize these applications with the strategy of mechanism including anti-cardiomyocyte death strategy, activation of neovascularization, antioxidants strategy, immunomodulation, anti-cardiac remodeling, and cardiac repair.


Subject(s)
Myocardial Infarction , Nanomedicine , Myocardial Infarction/therapy , Humans , Animals , Myocytes, Cardiac/drug effects , Antioxidants/therapeutic use , Nanostructures/therapeutic use , Nanostructures/chemistry , Neovascularization, Physiologic/drug effects
2.
Int J Nanomedicine ; 19: 6499-6513, 2024.
Article in English | MEDLINE | ID: mdl-38946887

ABSTRACT

Purpose: To address the problem of suboptimal reactive oxygen species (ROS) production in Radiation therapy (RT) which was resulted from exacerbated tumor hypoxia and the heterogeneous distribution of radiation sensitizers. Materials and Methods: In this work, a novel nanomedicine, designated as PLGA@IR780-Bi-DTPA (PIBD), was engineered by loading the radiation sensitizer Bi-DTPA and the photothermal agent IR780 onto poly(lactic-co-glycolic acid) (PLGA). This design leverages the tumor-targeting ability of IR780 to ensure selective accumulation of the nanoparticles in tumor cells, particularly within the mitochondria. The effect of the photothermal therapy-enhanced radiation therapy was also examined to assess the alleviation of hypoxia and the enhancement of radiation sensitivity. Results: The PIBD nanoparticles exhibited strong capacity in mitochondrial targeting and selective tumor accumulation. Upon activation by 808 nm laser irradiation, the nanoparticles effectively alleviated local hypoxia by photothermal effect enhanced blood supplying to improve oxygen content, thereby enhancing the ROS production for effective RT. Comparative studies revealed that PIBD-induced RT significantly outperformed conventional RT in treating hypoxic tumors. Conclusion: This design of tumor-targeting photothermal therapy-enhanced radiation therapy nanomedicine would advance the development of targeted drug delivery system for effective RT regardless of hypoxic microenvironment.


Subject(s)
Nanoparticles , Photothermal Therapy , Polylactic Acid-Polyglycolic Acid Copolymer , Reactive Oxygen Species , Animals , Photothermal Therapy/methods , Reactive Oxygen Species/metabolism , Nanoparticles/chemistry , Cell Line, Tumor , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Mice , Indoles/pharmacology , Indoles/chemistry , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/radiotherapy , Neoplasms/therapy , Neoplasms/metabolism , Nanomedicine
3.
Theranostics ; 14(9): 3486-3508, 2024.
Article in English | MEDLINE | ID: mdl-38948064

ABSTRACT

Rationale: Device implantation frequently triggers cardiac remodeling and fibrosis, with monocyte-driven inflammatory responses precipitating arrhythmias. This study investigates the role of m6A modification enzymes METTL3 and METTL14 in these responses and explores a novel therapeutic strategy targeting these modifications to mitigate cardiac remodeling and fibrosis. Methods: Peripheral blood mononuclear cells (PBMCs) were collected from patients with ventricular septal defects (VSD) who developed conduction blocks post-occluder implantation. The expression of METTL3 and METTL14 in PBMCs was measured. METTL3 and METTL14 deficiencies were induced to evaluate their effect on angiotensin II (Ang II)-induced myocardial inflammation and fibrosis. m6A modifications were analyzed using methylated RNA immunoprecipitation followed by quantitative PCR. NF-κB pathway activity and levels of monocyte migration and fibrogenesis markers (CXCR2 and TGF-ß1) were assessed. An erythrocyte microvesicle-based nanomedicine delivery system was developed to target activated monocytes, utilizing the METTL3 inhibitor STM2457. Cardiac function was evaluated via echocardiography. Results: Significant upregulation of METTL3 and METTL14 was observed in PBMCs from patients with VSD occluder implantation-associated persistent conduction block. Deficiencies in METTL3 and METTL14 significantly reduced Ang II-induced myocardial inflammation and fibrosis by decreasing m6A modification on MyD88 and TGF-ß1 mRNAs. This disruption reduced NF-κB pathway activation, lowered CXCR2 and TGF-ß1 levels, attenuated monocyte migration and fibrogenesis, and alleviated cardiac remodeling. The erythrocyte microvesicle-based nanomedicine delivery system effectively targeted inflamed cardiac tissue, reducing inflammation and fibrosis and improving cardiac function. Conclusion: Inhibiting METTL3 and METTL14 in monocytes disrupts the NF-κB feedback loop, decreases monocyte migration and fibrogenesis, and improves cardiac function. Targeting m6A modifications of monocytes with STM2457, delivered via erythrocyte microvesicles, reduces inflammation and fibrosis, offering a promising therapeutic strategy for cardiac remodeling associated with device implantation.


Subject(s)
Fibrosis , Methyltransferases , Monocytes , NF-kappa B , Humans , Methyltransferases/metabolism , Methyltransferases/genetics , Monocytes/metabolism , Male , Animals , NF-kappa B/metabolism , Erythrocytes/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Female , Methylation , Mice , Transforming Growth Factor beta1/metabolism , Cell-Derived Microparticles/metabolism , Leukocytes, Mononuclear/metabolism , Angiotensin II/metabolism , Receptors, Interleukin-8B/metabolism , Receptors, Interleukin-8B/genetics , Ventricular Remodeling , Myocardium/metabolism , Myocardium/pathology , Nanomedicine/methods
4.
Article in English | MEDLINE | ID: mdl-38955512

ABSTRACT

Enzyme-based therapy has garnered significant attention for its current applications in various diseases. Despite the notable advantages associated with the use of enzymes as therapeutic agents, that could have high selectivity, affinity, and specificity for the target, their application faces challenges linked to physico-chemical and pharmacological properties. These limitations can be addressed through the encapsulation of enzymes in nanoplatforms as a comprehensive solution to mitigate their degradation, loss of activity, off-target accumulation, and immunogenicity, thus enhancing bioavailability, therapeutic efficacy, and circulation time, thereby reducing the number of administrations, and ameliorating patient compliance. The exploration of novel nanomedicine-based enzyme therapeutics for the treatment of challenging diseases stands as a paramount goal in the contemporary scientific landscape, but even then it is often not enough. Combining an enzyme with another therapeutic (e.g., a small molecule, another enzyme or protein, a monoclonal antibody, or a nucleic acid) within a single nanocarrier provides innovative multidrug-integrated therapy and ensures that both the actives arrive at the target site and exert their therapeutic effect, leading to synergistic action and superior therapeutic efficacy. Moreover, this strategic approach could be extended to gene therapy, a field that nowadays has gained increasing attention, as enzymes acting at genomic level and nucleic acids may be combined for synergistic therapy. This multicomponent therapeutic approach opens opportunities for promising future developments. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.


Subject(s)
Enzyme Therapy , Nanomedicine , Humans , Animals
5.
AAPS J ; 26(4): 74, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38955936

ABSTRACT

The paper highlights the necessity for a robust regulatory framework for assessing nanomedicines and their off-patent counterparts, termed as nanosimilar, which could be considered as 'similar' to the prototype nanomedicine,based on essential criteria describing the 'similarity'. The term 'similarity' should be focused on criteria that describe nanocarriers, encompassing their physicochemical, thermodynamic, morphological, and biological properties, including surface interactions and pharmacokinetics. Nanocarriers can be regarded as advanced self-assembled excipients (ASAEs) due to their complexity and chaotic behavior and should be evaluated by using essential criteria in order for off-patent nanomedicines be termed as nanosimilars, from a regulatory perspective. Collaboration between the pharmaceutical industry, regulatory bodies, and artificial intelligence (AI) startups is pivotal for the precise characterization and approval processes for nanomedicines and nanosimilars and embracing innovative tools and terminology facilitates the development of a sustainable regulatory framework, ensuring safety and efficacy. This crucial shift toward precision R&D practices addresses the complexity inherent in nanocarriers, paving the way for therapeutic advancements with economic benefits.


Subject(s)
Nanomedicine , Nanomedicine/legislation & jurisprudence , Nanomedicine/methods , Humans , Biosimilar Pharmaceuticals/administration & dosage , Biosimilar Pharmaceuticals/pharmacokinetics , Artificial Intelligence , Nanoparticles , Drug Industry/legislation & jurisprudence , Drug Approval/legislation & jurisprudence , Drug Carriers/chemistry
6.
Nanotheranostics ; 8(4): 473-496, 2024.
Article in English | MEDLINE | ID: mdl-38961885

ABSTRACT

Cardiotoxicity, the often-overlooked second leading cause of death in cancer patients, has been associated with certain anticancer drugs. These drugs can induce cardiac damage through various pathways, and their adverse effects on the heart are not fully understood. Cardiotoxicity is a major issue in cancer treatment, particularly with chemotherapeutics, because it can cause cardiac dysfunction such as hypotension, heart failure, and even death. Doxorubicin, 5-fluorouracil, and trastuzumab, all of which are very potent anticancer drugs, are known to cause cardiotoxicity. When it comes to lowering cardiotoxicity and alleviating the harmful effects of chemotherapy medications, nanomedicine has the potential to transport therapeutic molecules. Nanotheranostics offers novel options for identifying and treating cardiotoxicity resulting from a wide range of substances, including anticancer medications. Additionally, theranostics platforms such as micellar systems, carbon-based nanomedicine, solid lipid nanoparticles, polymeric nanoparticles, and liposomes can transport chemotherapeutic medications while minimising their cardiotoxicity. The present level of understanding of the molecular and cellular processes that lead to cardiotoxicity in reaction to both traditional chemotherapy and targeted drug delivery systems is summarised in this article. This review delves into nanomedicine and nanotheranostics, with an emphasis on reducing anticancer medication-induced cardiac toxicity. Nanotheranostics provide potential solutions for early diagnosis and tailored therapy of heart injury by combining diagnostic and therapeutic capabilities into nanomedicine.


Subject(s)
Antineoplastic Agents , Cardiotoxicity , Nanomedicine , Theranostic Nanomedicine , Humans , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Cardiotoxicity/etiology , Nanomedicine/methods , Theranostic Nanomedicine/methods , Animals , Heart Diseases/chemically induced , Neoplasms/drug therapy , Nanoparticles/chemistry
7.
Int J Nanomedicine ; 19: 6295-6317, 2024.
Article in English | MEDLINE | ID: mdl-38919774

ABSTRACT

Nanoscale metal-organic frameworks (MOFs) offer high biocompatibility, nanomaterial permeability, substantial specific surface area, and well-defined pores. These properties make MOFs valuable in biomedical applications, including biological targeting and drug delivery. They also play a critical role in tumor diagnosis and treatment, including tumor cell targeting, identification, imaging, and therapeutic methods such as drug delivery, photothermal effects, photodynamic therapy, and immunogenic cell death. The diversity of MOFs with different metal centers, organics, and surface modifications underscores their multifaceted contributions to tumor research and treatment. This review is a summary of these roles and mechanisms. The final section of this review summarizes the current state of the field and discusses prospects that may bring MOFs closer to pharmaceutical applications.


Subject(s)
Metal-Organic Frameworks , Nanocomposites , Neoplasms , Metal-Organic Frameworks/chemistry , Humans , Neoplasms/diagnostic imaging , Neoplasms/therapy , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Drug Delivery Systems/methods , Animals , Photochemotherapy/methods , Antineoplastic Agents/chemistry , Nanomedicine/methods
8.
J Nanobiotechnology ; 22(1): 365, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918839

ABSTRACT

Bacteriophages (phages) represent a unique category of viruses with a remarkable ability to selectively infect host bacteria, characterized by their assembly from proteins and nucleic acids. Leveraging their exceptional biological properties and modifiable characteristics, phages emerge as innovative, safe, and efficient delivery vectors. The potential drawbacks associated with conventional nanocarriers in the realms of drug and gene delivery include a lack of cell-specific targeting, cytotoxicity, and diminished in vivo transfection efficiency. In contrast, engineered phages, when employed as cargo delivery vectors, hold the promise to surmount these limitations and attain enhanced delivery efficacy. This review comprehensively outlines current strategies for the engineering of phages, delineates the principal types of phages utilized as nanocarriers in drug and gene delivery, and explores the application of phage-based delivery systems in disease therapy. Additionally, an incisive analysis is provided, critically examining the challenges confronted by phage-based delivery systems within the domain of nanotechnology. The primary objective of this article is to furnish a theoretical reference that contributes to the reasoned design and development of potent phage-based delivery systems.


Subject(s)
Bacteriophages , Drug Delivery Systems , Nanomedicine , Bacteriophages/genetics , Humans , Nanomedicine/methods , Drug Delivery Systems/methods , Animals , Gene Transfer Techniques , Drug Carriers/chemistry , Nanoparticles/chemistry , Nanotechnology/methods
10.
AAPS PharmSciTech ; 25(6): 140, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890191

ABSTRACT

Nanotechnology has significantly transformed cancer treatment by introducing innovative methods for delivering drugs effectively. This literature review provided an in-depth analysis of the role of nanocarriers in cancer therapy, with a particular focus on the critical concept of the 'stealth effect.' The stealth effect refers to the ability of nanocarriers to evade the immune system and overcome physiological barriers. The review investigated the design and composition of various nanocarriers, such as liposomes, micelles, and inorganic nanoparticles, highlighting the importance of surface modifications and functionalization. The complex interaction between the immune system, opsonization, phagocytosis, and the protein corona was examined to understand the stealth effect. The review carefully evaluated strategies to enhance the stealth effect, including surface coating with polymers, biomimetic camouflage, and targeting ligands. The in vivo behavior of stealth nanocarriers and their impact on pharmacokinetics, biodistribution, and toxicity were also systematically examined. Additionally, the review presented clinical applications, case studies of approved nanocarrier-based cancer therapies, and emerging formulations in clinical trials. Future directions and obstacles in the field, such as advancements in nanocarrier engineering, personalized nanomedicine, regulatory considerations, and ethical implications, were discussed in detail. The review concluded by summarizing key findings and emphasizing the transformative potential of stealth nanocarriers in revolutionizing cancer therapy. This review enhanced the comprehension of nanocarrier-based cancer therapies and their potential impact by providing insights into advanced studies, clinical applications, and regulatory considerations.


Subject(s)
Antineoplastic Agents , Drug Carriers , Nanoparticles , Neoplasms , Humans , Neoplasms/drug therapy , Drug Carriers/chemistry , Nanoparticles/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Animals , Drug Delivery Systems/methods , Nanomedicine/methods , Liposomes , Micelles , Tissue Distribution
11.
Int J Nanomedicine ; 19: 6099-6126, 2024.
Article in English | MEDLINE | ID: mdl-38911500

ABSTRACT

The relentless pursuit of effective cancer diagnosis and treatment strategies has led to the rapidly expanding field of nanotechnology, with a specific focus on nanocomposites. Nanocomposites, a combination of nanomaterials with diverse properties, have emerged as versatile tools in oncology, offering multifunctional platforms for targeted delivery, imaging, and therapeutic interventions. Nanocomposites exhibit great potential for early detection and accurate imaging in cancer diagnosis. Integrating various imaging modalities, such as magnetic resonance imaging (MRI), computed tomography (CT), and fluorescence imaging, into nanocomposites enables the development of contrast agents with enhanced sensitivity and specificity. Moreover, functionalizing nanocomposites with targeting ligands ensures selective accumulation in tumor tissues, facilitating precise imaging and diagnostic accuracy. On the therapeutic front, nanocomposites have revolutionized cancer treatment by overcoming traditional challenges associated with drug delivery. The controlled release of therapeutic agents from nanocomposite carriers enhances drug bioavailability, reduces systemic toxicity, and improves overall treatment efficacy. Additionally, the integration of stimuli-responsive components within nanocomposites enables site-specific drug release triggered by the unique microenvironment of the tumor. Despite the remarkable progress in the field, challenges such as biocompatibility, scalability, and long-term safety profiles remain. This article provides a comprehensive overview of recent developments, challenges, and prospects, emphasizing the transformative potential of nanocomposites in revolutionizing the landscape of cancer diagnostics and therapeutics. In Conclusion, integrating nanocomposites in cancer diagnosis and treatment heralds a new era for precision medicine.


Subject(s)
Nanocomposites , Neoplasms , Humans , Nanocomposites/chemistry , Neoplasms/diagnostic imaging , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/therapy , Animals , Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Magnetic Resonance Imaging/methods , Contrast Media/chemistry , Nanomedicine/methods , Tomography, X-Ray Computed , Drug Carriers/chemistry
12.
Nanoscale ; 16(25): 11969-11976, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38874918

ABSTRACT

Nanomedicine is a field at the intersection of nanotechnology and medicine, promising due to its potential to revolutionize healthcare. Despite its long trajectory, there is still a long road ahead for its full development, and smart design of nanomedicines is still a challenge. Among other problems, this is due to the scarcity of tools available for the precise visualization and comprehension of nano-bio interactions, impeding progress towards the clinical phase. One of the developed tools that stands out to be a strong nanoscopy technique for studying nano-delivery systems within cellular environments is expansion microscopy (ExM). This technique was used for tissue and cell expansion and most recently for lipid molecule expansion inside cells. Herein, we present for the first time polyplex expansion microscopy (PExM); a comprehensive examination of ExM as an already developed technique, but adapted for expanding polymer based nanocarriers, in particular polyplexes within cells, allowing the analysis of their trafficking. With our method set up, PExM will be extensively used for the study of polyplex nanoparticle cell trafficking, becoming a high-resolution technique which can also be applied to primary amine containing polymeric nanoparticles without requiring expensive super-resolution microscopes.


Subject(s)
Nanoparticles , Humans , Nanoparticles/chemistry , Polymers/chemistry , Microscopy/methods , Nanomedicine
13.
J Nanobiotechnology ; 22(1): 354, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902775

ABSTRACT

Fundus neovascularization diseases are a series of blinding eye diseases that seriously impair vision worldwide. Currently, the means of treating these diseases in clinical practice are continuously evolving and have rapidly revolutionized treatment opinions. However, key issues such as inadequate treatment effectiveness, high rates of recurrence, and poor patient compliance still need to be urgently addressed. Multifunctional nanomedicine can specifically respond to both endogenous and exogenous microenvironments, effectively deliver drugs to specific targets and participate in activities such as biological imaging and the detection of small molecules. Nano-in-micro (NIM) delivery systems such as metal, metal oxide and up-conversion nanoparticles (NPs), quantum dots, and carbon materials, have shown certain advantages in overcoming the presence of physiological barriers within the eyeball and are widely used in the treatment of ophthalmic diseases. Few studies, however, have evaluated the efficacy of NIM delivery systems in treating fundus neovascular diseases (FNDs). The present study describes the main clinical treatment strategies and the adverse events associated with the treatment of FNDs with NIM delivery systems and summarizes the anatomical obstacles that must be overcome. In this review, we wish to highlight the principle of intraocular microenvironment normalization, aiming to provide a more rational approach for designing new NIM delivery systems to treat specific FNDs.


Subject(s)
Drug Delivery Systems , Humans , Animals , Drug Delivery Systems/methods , Neovascularization, Pathologic/drug therapy , Fundus Oculi , Quantum Dots/chemistry , Multifunctional Nanoparticles/chemistry , Retinal Neovascularization/drug therapy , Nanomedicine/methods , Nanoparticles/chemistry
14.
J Nanobiotechnology ; 22(1): 364, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38915007

ABSTRACT

Photothermal therapy (PTT) is a promising cancer treatment method due to its ability to induce tumor-specific T cell responses and enhance therapeutic outcomes. However, incomplete PTT can leave residual tumors that often lead to new metastases and decreased patient survival in clinical scenarios. This is primarily due to the release of ATP, a damage-associated molecular pattern that quickly transforms into the immunosuppressive metabolite adenosine by CD39, prevalent in the tumor microenvironment, thus promoting tumor immune evasion. This study presents a photothermal nanomedicine fabricated by electrostatic adsorption among the Fe-doped polydiaminopyridine (Fe-PDAP), indocyanine green (ICG), and CD39 inhibitor sodium polyoxotungstate (POM-1). The constructed Fe-PDAP@ICG@POM-1 (FIP) can induce tumor PTT and immunogenic cell death when exposed to a near-infrared laser. Significantly, it can inhibit the ATP-adenosine pathway by dual-directional immunometabolic regulation, resulting in increased ATP levels and decreased adenosine synthesis, which ultimately reverses the immunosuppressive microenvironment and increases the susceptibility of immune checkpoint blockade (aPD-1) therapy. With the aid of aPD-1, the dual-directional immunometabolic regulation strategy mediated by FIP can effectively suppress/eradicate primary and distant tumors and evoke long-term solid immunological memory. This study presents an immunometabolic control strategy to offer a salvage option for treating residual tumors following incomplete PTT.


Subject(s)
Immunotherapy , Nanomedicine , Photothermal Therapy , Tumor Microenvironment , Animals , Photothermal Therapy/methods , Immunotherapy/methods , Mice , Nanomedicine/methods , Tumor Microenvironment/drug effects , Cell Line, Tumor , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Neoplasms/therapy , Adenosine Triphosphate/metabolism , Adenosine/pharmacology , Adenosine/chemistry , Mice, Inbred C57BL , Apyrase/metabolism , Female , Phototherapy/methods
15.
Biomed Pharmacother ; 176: 116904, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38878686

ABSTRACT

Globally, cancer is a serious health problem. It is unfortunate that current anti-cancer strategies are insufficiently specific and damage the normal tissues. There's urgent need for development of new anti-cancer strategies. More recently, increasing attention has been paid to the new application of ferroptosis and nano materials in cancer research. Ferroptosis, a condition characterized by excessive reactive oxygen species-induced lipid peroxidation, as a new programmed cell death mode, exists in the process of a number of diseases, including cancers, neurodegenerative disease, cerebral hemorrhage, liver disease, and renal failure. There is growing evidence that inducing ferroptosis has proven to be an effective strategy against a variety of chemo-resistant cancer cells. Nano-drug delivery system based on nanotechnology provides a highly promising platform with the benefits of precise control of drug release and reduced toxicity and side effects. This paper reviews the latest advances of combination therapy strategies based on biomedical nanotechnology induced ferroptosis for cancer therapeutics. Given the new chances and challenges in this emerging area, we need more attention to the combination of nanotechnology and ferroptosis in the treatment of cancer in the future.


Subject(s)
Ferroptosis , Neoplasms , Ferroptosis/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Nanoparticles , Nanotechnology/methods , Nanoparticle Drug Delivery System , Drug Delivery Systems/methods , Combined Modality Therapy , Reactive Oxygen Species/metabolism , Nanomedicine/methods
17.
Int J Mol Sci ; 25(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38892023

ABSTRACT

We launched this Special Issue amidst the COVID-19 pandemic, spurred by the growing interest in nanotherapeutic formulations for delivering SARS-CoV-2 viral messenger Ribonucleic Acid (mRNA) vaccines [...].


Subject(s)
COVID-19 Vaccines , COVID-19 , Nanomedicine , SARS-CoV-2 , Humans , Nanomedicine/methods , COVID-19/virology , COVID-19/prevention & control
18.
Molecules ; 29(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38893373

ABSTRACT

Developing clinically meaningful nanomedicines for cancer therapy requires the drugs to be effective, safe, simple, cheap, and easy to store. In the present work, we report that a simple cationic Fe(III)-rich salt of [FeIIICl(TMPPH2)][FeIIICl4]2 (Fe-TMPP) exhibits a superior anticancer performance on a broad spectrum of cancer cell lines, including breast, colorectal cancer, liver, pancreatic, prostate, and gastric cancers, with half maximal inhibitory concentration (IC50) values in the range of 0.098-3.97 µM (0.066-2.68 µg mL-1), comparable to the best-reported medicines. Fe-TMPP can form stand-alone nanoparticles in water without the need for extra surface modification or organic-solvent-assisted antisolvent precipitation. Critically, Fe-TMPP is TME-responsive (TME = tumor microenvironment), and can only elicit its function in the TME with overexpressed H2O2, converting H2O2 to the cytotoxic •OH to oxidize the phospholipid of the cancer cell membrane, causing ferroptosis, a programmed cell death process of cancer cells.


Subject(s)
Antineoplastic Agents , Ferroptosis , Nanomedicine , Humans , Ferroptosis/drug effects , Cell Line, Tumor , Nanomedicine/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Ferric Compounds/chemistry , Tumor Microenvironment/drug effects , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/pharmacology , Cell Survival/drug effects , Neoplasms/drug therapy , Neoplasms/pathology
19.
Molecules ; 29(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38893460

ABSTRACT

There is a myriad of diseases that plague the world ranging from infectious, cancer and other chronic diseases with varying interventions. However, the dynamism of causative agents of infectious diseases and incessant mutations accompanying other forms of chronic diseases like cancer, have worsened the treatment outcomes. These factors often lead to treatment failure via different drug resistance mechanisms. More so, the cost of developing newer drugs is huge. This underscores the need for a paradigm shift in the drug delivery approach in order to achieve desired treatment outcomes. There is intensified research in nanomedicine, which has shown promises in improving the therapeutic outcome of drugs at preclinical stages with increased efficacy and reduced toxicity. Regardless of the huge benefits of nanotechnology in drug delivery, challenges such as regulatory approval, scalability, cost implication and potential toxicity must be addressed via streamlining of regulatory hurdles and increased research funding. In conclusion, the idea of nanotechnology in drug delivery holds immense promise for optimizing therapeutic outcomes. This work presents opportunities to revolutionize treatment strategies, providing expert opinions on translating the huge amount of research in nanomedicine into clinical benefits for patients with resistant infections and cancer.


Subject(s)
Drug Delivery Systems , Nanomedicine , Nanostructures , Humans , Nanostructures/chemistry , Nanomedicine/methods , Neoplasms/drug therapy , Animals , Nanotechnology/methods
20.
ACS Appl Mater Interfaces ; 16(23): 29844-29855, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38829261

ABSTRACT

Copper plays critical roles as a metal active site cofactor and metalloallosteric signal for enzymes involved in cell proliferation and metabolism, making it an attractive target for cancer therapy. In this study, we investigated the efficacy of polydopamine nanoparticles (PDA NPs), classically applied for metal removal from water, as a therapeutic strategy for depleting intracellular labile copper pools in triple-negative breast cancer models through the metal-chelating groups present on the PDA surface. By using the activity-based sensing probe FCP-1, we could track the PDA-induced labile copper depletion while leaving total copper levels unchanged and link it to the selective MDA-MB-231 cell death. Further mechanistic investigations revealed that PDA NPs increased reactive oxygen species (ROS) levels, potentially through the inactivation of superoxide dismutase 1 (SOD1), a copper-dependent antioxidant enzyme. Additionally, PDA NPs were found to interact with the mitochondrial membrane, resulting in an increase in the mitochondrial membrane potential, which may contribute to enhanced ROS production. We employed an in vivo tumor model to validate the therapeutic efficacy of PDA NPs. Remarkably, in the absence of any additional treatment, the presence of PDA NPs alone led to a significant reduction in tumor volume by a factor of 1.66 after 22 days of tumor growth. Our findings highlight the potential of PDA NPs as a promising therapeutic approach for selectively targeting cancer by modulating copper levels and inducing oxidative stress, leading to tumor growth inhibition as shown in these triple-negative breast cancer models.


Subject(s)
Copper , Indoles , Nanoparticles , Polymers , Reactive Oxygen Species , Triple Negative Breast Neoplasms , Copper/chemistry , Copper/pharmacology , Polymers/chemistry , Polymers/pharmacology , Indoles/chemistry , Indoles/pharmacology , Humans , Animals , Mice , Nanoparticles/chemistry , Female , Reactive Oxygen Species/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Oxidation-Reduction , Nanomedicine , Cell Proliferation/drug effects , Homeostasis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Superoxide Dismutase-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...