Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.754
Filter
1.
Int J Nanomedicine ; 19: 6485-6497, 2024.
Article in English | MEDLINE | ID: mdl-38946886

ABSTRACT

Angiogenesis is a physiological process of forming new blood vessels that has pathological importance in seemingly unrelated illnesses like cancer, diabetes, and various inflammatory diseases. Treatment targeting angiogenesis has shown promise for these types of diseases, but current anti-angiogenic agents have critical limitations in delivery and side-effects. This necessitates exploration of alternative approaches like biomolecule-based drugs. Proteins, lipids, and oligonucleotides have recently become popular in biomedicine, specifically as biocompatible components of therapeutic drugs. Their excellent bioavailability and potential bioactive and immunogenic properties make them prime candidates for drug discovery or drug delivery systems. Lipid-based liposomes have become standard vehicles for targeted nanoparticle (NP) delivery, while protein and nucleotide NPs show promise for environment-sensitive delivery as smart NPs. Their therapeutic applications have initially been hampered by short circulation times and difficulty of fabrication but recent developments in nanofabrication and NP engineering have found ways to circumvent these disadvantages, vastly improving the practicality of biomolecular NPs. In this review, we are going to briefly discuss how biomolecule-based NPs have improved anti-angiogenesis-based therapy.


Subject(s)
Angiogenesis Inhibitors , Neovascularization, Pathologic , Theranostic Nanomedicine , Humans , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/administration & dosage , Theranostic Nanomedicine/methods , Neovascularization, Pathologic/drug therapy , Animals , Liposomes/chemistry , Nanostructures/chemistry , Neoplasms/drug therapy , Drug Delivery Systems/methods , Oligonucleotides/chemistry , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacokinetics , Oligonucleotides/pharmacology , Proteins/chemistry , Proteins/administration & dosage , Lipids/chemistry , Nanoparticles/chemistry
2.
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
3.
Theranostics ; 14(10): 3900-3908, 2024.
Article in English | MEDLINE | ID: mdl-38994024

ABSTRACT

Background: Osteoarthritis (OA) standing as the most prevalent form of arthritis, closely associates with heightened levels of reactive oxygen species, particularly hypochlorous acid (HOCl). Although there are numerous probes available for detecting HOCl in the OA region, probes with dual functions of diagnostic and therapeutic capabilities are still significantly lacking. While this type of probe can reduce the time gap between diagnosis and treatment, which is clinically needed. Methods: We developed a fluorescent probe (DHU-CBA1) toward HOCl with theranostics functions through the release of methylene blue (MB) and ibuprofen (IBP) in this work. DHU-CBA1 can detect HOCl with high specificity and sensitivity, releasing MB and IBP with an impressive efficiency of ≥ 95% in vitro. Results: DHU-CBA1 exhibits good biosafety, enabling in vivo imaging of endogenous HOCl, along with reducing arthritis scores, improving synovitis and cartilage damage, and maintaining catabolic balance while alleviating senescence in cartilage. Conclusions: This study proposes a novel approach to enhance osteoarthritis therapy by releasing IBP via a smart HOCl-enabled fluorescent probe.


Subject(s)
Fluorescent Dyes , Hypochlorous Acid , Ibuprofen , Methylene Blue , Osteoarthritis , Osteoarthritis/drug therapy , Fluorescent Dyes/chemistry , Ibuprofen/administration & dosage , Animals , Methylene Blue/chemistry , Mice , Humans , Theranostic Nanomedicine/methods , Male , Optical Imaging/methods , Reactive Oxygen Species/metabolism
5.
Molecules ; 29(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38999115

ABSTRACT

According to the World Health Organization (WHO) and the International Agency for Research on Cancer (IARC), the number of cancer cases and deaths worldwide is predicted to nearly double by 2030, reaching 21.7 million cases and 13 million fatalities. The increase in cancer mortality is due to limitations in the diagnosis and treatment options that are currently available. The close relationship between diagnostics and medicine has made it possible for cancer patients to receive precise diagnoses and individualized care. This article discusses newly developed compounds with potential for photodynamic therapy and diagnostic applications, as well as those already in use. In addition, it discusses the use of artificial intelligence in the analysis of diagnostic images obtained using, among other things, theranostic agents.


Subject(s)
Artificial Intelligence , Neoplasms , Photochemotherapy , Humans , Neoplasms/drug therapy , Neoplasms/diagnosis , Neoplasms/therapy , Photochemotherapy/methods , Theranostic Nanomedicine/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
6.
Mikrochim Acta ; 191(7): 433, 2024 06 29.
Article in English | MEDLINE | ID: mdl-38951214

ABSTRACT

A cancer-targeted glutathione (GSH)-gated theranostic probe (CGT probe) for intracellular miRNA imaging and combined treatment of self-sufficient starvation therapy (ST) and chemodynamic therapy (CDT) was developed. The CGT probe is constructed using MnO2 nanosheet (MS) as carrier material to adsorb the elaborately designed functional DNAs. It can be internalized by cancer cells via specific recognition between the AS1411 aptamer and nucleolin. After CGT probe entering the cancer cells, the overexpressed GSH, as gate-control, can degrade MS to Mn2+ which can be used for CDT by Fenton-like reaction. Simultaneously, Mn2+-mediated CDT can further cascade with the enzyme-like activities (catalase-like activity and glucose oxidase-like activity) of CGT probe, achieving self-sufficient ST/CDT synergistic therapy. Meanwhile, the anchored DNAs are released, achieving in situ signal amplification via disubstituted-catalytic hairpin assembly (DCHA) and FRET (fluorescence resonance energy transfer) imaging of miR-21. The in vitro and in vivo experiments demonstrated that accurate and sensitive miRNA detection can be achieved using the CGT probe. Overall, the ingenious CGT probe opens a new avenue for the development of early clinical diagnosis and cancer therapy.


Subject(s)
Fluorescence Resonance Energy Transfer , Glutathione , Manganese Compounds , MicroRNAs , Oxides , Humans , Glutathione/chemistry , Glutathione/metabolism , Animals , Manganese Compounds/chemistry , Oxides/chemistry , Aptamers, Nucleotide/chemistry , Mice , Mice, Nude , Theranostic Nanomedicine/methods , Nucleolin , Neoplasms/diagnostic imaging , Nanostructures/chemistry , Oligodeoxyribonucleotides/chemistry , Mice, Inbred BALB C , Fluorescent Dyes/chemistry
7.
Med Oncol ; 41(8): 205, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39037549

ABSTRACT

In this study, we developed new gadolinium-graphene quantum dot nanoparticles (Gd-GQDs) as a theranostic platform for magnetic resonance imaging and improved the efficiency of radiotherapy in HPV-positive oropharyngeal cancer. Based on cell toxicity results, Gd-GQD NPs were nontoxic for both cancer and normal cell lines up to 25 µg/ml. These NPs enhance the cytotoxic effect of radiation only on cancer cells but not on normal cells. The flow cytometry analysis indicated that cell death mainly occurred in the late phase of apoptosis. The immunocytochemical analysis was used to evaluate apoptosis pathway proteins. The Bcl-2 and p53 protein levels did not differ statistically significantly between radiation alone group and those that received irradiation in combination with NPs. In contrast, the combination group exhibited a significant increase in Bax protein expression, suggesting that cells could undergo apoptosis independent of the p53 pathway. Magnetic resonance (MR) imaging showed that Gd-GQD NPs, when used at low concentrations, enhanced T1-weighted signal intensity resulting from T1 shortening effects. At higher concentrations, the T2 shortening effect became predominant and was able to decrease the signal intensity. Gd-GQD appears to offer a novel approach for enhancing the effectiveness of radiation treatment and facilitating MR imaging for monitoring HPV-positive tumors.


Subject(s)
Gadolinium , Magnetic Resonance Imaging , Oropharyngeal Neoplasms , Papillomavirus Infections , Quantum Dots , Humans , Oropharyngeal Neoplasms/virology , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/therapy , Oropharyngeal Neoplasms/radiotherapy , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Magnetic Resonance Imaging/methods , Apoptosis/drug effects , Theranostic Nanomedicine/methods , Cell Line, Tumor
8.
Theranostics ; 14(9): 3634-3652, 2024.
Article in English | MEDLINE | ID: mdl-38948059

ABSTRACT

Rationale: Molecular imaging of microenvironment by hypoxia-activatable fluorescence probes has emerged as an attractive approach to tumor diagnosis and image-guided treatment. Difficulties remain in its translational applications due to hypoxia heterogeneity in tumor microenvironments, making it challenging to image hypoxia as a reliable proxy of tumor distribution. Methods: We report a modularized theranostics platform to fluorescently visualize hypoxia via light-modulated signal compensation to overcome tumor heterogeneity, thereby serving as a diagnostic tool for image-guided surgical resection and photodynamic therapy. Specifically, the platform integrating dual modules of fluorescence indicator and photodynamic moderator using supramolecular host-guest self-assembly, which operates cooperatively as a cascaded "AND" logic gate. First, tumor enrichment and specific fluorescence turn-on in hypoxic regions were accessible via tumor receptors and cascaded microenvironment signals as simultaneous inputs of the "AND" gate. Second, image guidance by a lighted fluorescence module and light-mediated endogenous oxygen consumption of a photodynamic module as dual inputs of "AND" gate collaboratively enabled light-modulated signal compensation in situ, indicating homogeneity of enhanced hypoxia-related fluorescence signals throughout a tumor. Results: In in vitro and in vivo analyses, the biocompatible platform demonstrated several strengths including a capacity for dual tumor targeting to progressively facilitate specific fluorescence turn-on, selective signal compensation, imaging-time window extension conducive to precise normalized image-guided treatment, and the functionality of tumor glutathione depletion to improve photodynamic efficacy. Conclusion: The hypoxia-activatable, image-guided theranostic platform demonstrated excellent potential for overcoming hypoxia heterogeneity in tumors.


Subject(s)
Optical Imaging , Theranostic Nanomedicine , Animals , Theranostic Nanomedicine/methods , Humans , Optical Imaging/methods , Mice , Tumor Microenvironment , Cell Line, Tumor , Fluorescent Dyes/chemistry , Photochemotherapy/methods , Neoplasms/diagnostic imaging , Neoplasms/therapy , Mice, Nude , Surgery, Computer-Assisted/methods
9.
Molecules ; 29(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38930939

ABSTRACT

Atherosclerosis continues to be a leading cause of morbidity and mortality globally. The precise evaluation of the extent of an atherosclerotic plaque is essential for forecasting its likelihood of causing health concerns and tracking treatment outcomes. When compared to conventional methods used, nanoparticles offer clear benefits and excellent development opportunities for the detection and characterisation of susceptible atherosclerotic plaques. In this review, we analyse the recent advancements of nanoparticles as theranostics in the management of atherosclerosis, with an emphasis on applications in drug delivery. Furthermore, the main issues that must be resolved in order to advance clinical utility and future developments of NP research are discussed. It is anticipated that medical NPs will develop into complex and advanced next-generation nanobotics that can carry out a variety of functions in the bloodstream.


Subject(s)
Atherosclerosis , Drug Delivery Systems , Nanoparticles , Humans , Atherosclerosis/drug therapy , Nanoparticles/chemistry , Drug Delivery Systems/methods , Animals , Theranostic Nanomedicine/methods , Plaque, Atherosclerotic/drug therapy , Drug Carriers/chemistry
10.
J Mater Sci Mater Med ; 35(1): 32, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896160

ABSTRACT

This study leverages nanotechnology by encapsulating indocyanine green (ICG) and paclitaxel (Tax) using zeolitic imidazolate frameworks-8 (ZIF-8) as a scaffold. This study aims to investigate the chemo-photothermal therapeutic potential of ZIF-8@ICG@Tax nanoparticles (NPs) in the treatment of non-small cell lung cancer (NSCLC). An "all-in-one" theranostic ZIF-8@ICG@Tax NPs was conducted by self-assembly based on electrostatic interaction. First, the photothermal effect, stability, pH responsiveness, drug release, and blood compatibility of ZIF-8@ICG@Tax were evaluated through in vitro testing. Furthermore, the hepatic and renal toxicity of ZIF-8@ICG@Tax were assessed through in vivo testing. Additionally, the anticancer effects of these nanoparticles were investigated both in vitro and in vivo. Uniform and stable chemo-photothermal ZIF-8@ICG@Tax NPs had been successfully synthesized and had outstanding drug releasing capacities. Moreover, ZIF-8@ICG@Tax NPs showed remarkable responsiveness dependent both on pH in the tumor microenvironment and NIR irradiation, allowing for targeted drug delivery and controlled drug release. NIR irradiation can enhance the tumor cell response to ZIF-8@ICG@Tax uptake, thereby promoting the anti-tumor growth in vitro and in vivo. ZIF-8@ICG@Tax and NIR irradiation have demonstrated remarkable synergistic anti-tumor growth properties compared to their individual components. This novel theranostic chemo-photothermal NPs hold great potential as a viable treatment option for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug Liberation , Indocyanine Green , Lung Neoplasms , Nanoparticles , Paclitaxel , Theranostic Nanomedicine , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Indocyanine Green/chemistry , Humans , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Hydrogen-Ion Concentration , Nanoparticles/chemistry , Theranostic Nanomedicine/methods , Paclitaxel/chemistry , Paclitaxel/pharmacology , Mice , Zeolites/chemistry , Infrared Rays , Phototherapy/methods , Mice, Inbred BALB C , Cell Line, Tumor , A549 Cells , Metal-Organic Frameworks/chemistry , Mice, Nude , Drug Delivery Systems , Imidazoles
11.
Int J Nanomedicine ; 19: 5479-5492, 2024.
Article in English | MEDLINE | ID: mdl-38863646

ABSTRACT

Background: In recent years, PD-L1 has been primarily utilized as an immune checkpoint marker in cancer immunotherapy. However, due to tumor heterogeneity, the response rate to such therapies often falls short of expectations. In addition to its role in immunotherapy, PD-L1 serves as a specific target on the surface of tumor cells for targeted diagnostic and therapeutic interventions. There is an absence of a fully developed PD-L1-targeted diagnostic and therapeutic probe for clinical use, which constrains the exploration and clinical exploitation of this target. Methods and Results: In this study, we engineered a PD-L1-targeted probe with multimodal imaging and dual therapeutic functionalities utilizing organic melanin nanoparticles. Functionalization with the WL12-SH peptide endowed the nanoprobe with specific targeting capabilities. Subsequent radiolabeling with 89Zr (half-life: 100.8 hours) and chelation of Mn2+ ions afforded the probe the capacity for simultaneous PET and MRI imaging modalities. Cellular uptake assays revealed pronounced specificity, with -positive cells exhibiting significantly higher uptake than -negative counterparts (p < 0.05). Dual-modal PET/MRI imaging delineated rapid and sustained accumulation at the neoplastic site, yielding tumor-to-non-tumor (T/NT) signal ratios at 24 hours post-injection of 16.67±3.45 for PET and 6.63±0.64 for MRI, respectively. We conjugated the therapeutic radionuclide 131I (half-life: 8.02 days) to the construct and combined low-dose radiotherapy and photothermal treatment (PTT), culminating in superior antitumor efficacy while preserving a high safety profile. The tumors in the cohort receiving the dual-modality therapy exhibited significantly reduced volume and weight compared to those in the control and monotherapy groups. Conclusion: We developed and applied a novel -targeted multimodal theranostic nanoprobe, characterized by its high specificity and superior imaging capabilities as demonstrated in PET/MRI modalities. Furthermore, this nanoprobe facilitates potent therapeutic efficacy at lower radionuclide doses when used in conjunction with PTT.


Subject(s)
B7-H1 Antigen , Magnetic Resonance Imaging , Multimodal Imaging , Nanoparticles , Positron-Emission Tomography , Theranostic Nanomedicine , Theranostic Nanomedicine/methods , Animals , B7-H1 Antigen/metabolism , Positron-Emission Tomography/methods , Nanoparticles/chemistry , Humans , Magnetic Resonance Imaging/methods , Multimodal Imaging/methods , Cell Line, Tumor , Mice , Melanins/chemistry , Zirconium/chemistry , Radioisotopes/chemistry , Female , Immunotherapy/methods
12.
J Control Release ; 371: 429-444, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849096

ABSTRACT

Protein-based nanoparticles have garnered significant attention in theranostic applications due to their superior biocompatibility, exceptional biodegradability and ease of functionality. Compared to other nanocarriers, protein-based nanoparticles offer additional advantages, including biofunctionality and precise molecular recognition abilities, which make them highly effective in navigating complex biological environments. Moreover, proteins can serve as powerful tools with self-assembling structures and reagents that enhance cell penetration. And their derivation from abundant renewable sources and ability to degrade into harmless amino acids further enhance their suitability for biomedical applications. However, protein-based nanoparticles have so far not realized their full potential. In this review, we summarize recent advances in the use of protein nanoparticles in tumor diagnosis and treatment and outline typical methods for preparing protein nanoparticles. The review of protein nanoparticles may provide useful new insights into the development of biomaterial fabrication.


Subject(s)
Drug Delivery Systems , Nanoparticles , Neoplasms , Proteins , Theranostic Nanomedicine , Humans , Neoplasms/drug therapy , Theranostic Nanomedicine/methods , Nanoparticles/chemistry , Animals , Proteins/administration & dosage , Proteins/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry
13.
Proc Natl Acad Sci U S A ; 121(25): e2322403121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38865273

ABSTRACT

Fluorine magnetic resonance imaging (19F-MRI) is particularly promising for biomedical applications owing to the absence of fluorine in most biological systems. However, its use has been limited by the lack of safe and water-soluble imaging agents with high fluorine contents and suitable relaxation properties. We report innovative 19F-MRI agents based on supramolecular dendrimers self-assembled by an amphiphilic dendrimer composed of a hydrophobic alkyl chain and a hydrophilic dendron. Specifically, this amphiphilic dendrimer bears multiple negatively charged terminals with high fluorine content, which effectively prevented intra- and intermolecular aggregation of fluorinated entities via electrostatic repulsion. This permitted high fluorine nuclei mobility alongside good water solubility with favorable relaxation properties for use in 19F-MRI. Importantly, the self-assembling 19F-MRI agent was able to encapsulate the near-infrared fluorescence (NIRF) agent DiR and the anticancer drug paclitaxel for multimodal 19F-MRI and NIRF imaging of and theranostics for pancreatic cancer, a deadly disease for which there remains no adequate early detection method or efficacious treatment. The 19F-MRI and multimodal 19F-MRI and NIRF imaging studies on human pancreatic cancer xenografts in mice confirmed the capability of both imaging modalities to specifically image the tumors and demonstrated the efficacy of the theranostic agent in cancer treatment, largely outperforming the clinical anticancer drug paclitaxel. Consequently, these dendrimer nanosystems constitute promising 19F-MRI agents for effective cancer management. This study offers a broad avenue to the construction of 19F-MRI agents and theranostics, exploiting self-assembling supramolecular dendrimer chemistry.


Subject(s)
Dendrimers , Fluorine , Theranostic Nanomedicine , Dendrimers/chemistry , Animals , Theranostic Nanomedicine/methods , Humans , Mice , Fluorine/chemistry , Paclitaxel/chemistry , Paclitaxel/therapeutic use , Magnetic Resonance Imaging/methods , Cell Line, Tumor , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/therapy , Fluorine-19 Magnetic Resonance Imaging/methods , Mice, Nude , Contrast Media/chemistry
14.
ACS Macro Lett ; 13(6): 768-774, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38829688

ABSTRACT

The low therapeutic efficacy and potential long-term toxicity of antitumor treatments seriously limit the clinical application of phototherapies. Herein, we develop a degradable phototheranostic nanoplatform for NIR-II fluorescence bioimaging-guided synergistic photothermal (PTT) and photodynamic therapies (PDT) and immune activation to inhibit tumor growth. The phototheranostic nanoplatform (CX@PSS) consists of multidisulfide-containing polyurethane loaded with a photosensitizer CX, which can be specifically degraded in the GSH overexpressed tumor microenvironment (TME) and exhibits good NIR-II fluorescence, photodynamic, and photothermal properties. Under 808 nm light irradiation, CX@PSS exhibits efficient photothermal conversion and ROS generation, which further induces immunogenic cell death (ICD), releasing tumor-associated antigens and activating the immune response. In vitro and in vivo studies confirm the potential of CX@PSS in NIR II FL imaging-guided tumor treatments by synergistic PTT, PDT, and immune activation. This work is expected to provide a new pathway for clinical applications of imaging-guided tumor diagnosis and treatments.


Subject(s)
Photochemotherapy , Photosensitizing Agents , Theranostic Nanomedicine , Tumor Microenvironment , Tumor Microenvironment/drug effects , Animals , Mice , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemistry , Theranostic Nanomedicine/methods , Photochemotherapy/methods , Optical Imaging/methods , Infrared Rays , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Cell Line, Tumor , Neoplasms/diagnostic imaging , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Photothermal Therapy/methods , Polyurethanes/chemistry , Polyurethanes/pharmacology
15.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892434

ABSTRACT

Many different types of nanoparticles have been suggested for tumor-targeted theranosis. However, most systems were prepared through a series of complicated processes and could not even overcome the blood-immune barriers. For the accurate diagnosis and effective treatment of cancers, herein we suggested the lipid micellar structure capturing quantum dot (QD) for cancer theranosis. The QD/lipid micelles (QDMs) were prepared using a simple self-assembly procedure and then conjugated with anti-epidermal growth factor receptor (EGFR) antibodies for tumor targeting. As a therapeutic agent, Bcl2 siRNA-cholesterol conjugates were loaded on the surface of QDMs. The EGFR-directed QDMs containing Bcl2 siRNA, so-called immuno-QDM/siBcl2 (iQDM/siBcl2), exhibited the more effective delivery of QDs and siBcl2 to target human colorectal cancer cells in cultures as well as in mouse xenografts. The effective in vivo targeting of iQDM/siBcl2 resulted in a more enhanced therapeutic efficacy of siBcl2 to the target cancer in mice. Based on the results, anti-EGFR QDM capturing therapeutic siRNA could be suggested as an alternative modality for tumor-targeted theranosis.


Subject(s)
ErbB Receptors , Proto-Oncogene Proteins c-bcl-2 , Quantum Dots , RNA, Small Interfering , Quantum Dots/chemistry , Animals , ErbB Receptors/genetics , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Humans , RNA, Small Interfering/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Mice , Cell Line, Tumor , Nanoparticles/chemistry , Lipids/chemistry , Theranostic Nanomedicine/methods , Xenograft Model Antitumor Assays , Micelles
16.
Int J Pharm ; 660: 124346, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38889853

ABSTRACT

Breast cancer, the second leading global cause of death, affects 2.1 million women annually, with an alarming 15 percent mortality rate. Among its diverse forms, Triple-negative breast cancer (TNBC) emerges as the deadliest, characterized by the absence of hormone receptors. This article underscores the urgent need for innovative treatment approaches in tackling TNBC, emphasizing the transformative potential of polymeric nanomaterials (PNMs). Evolved through nanotechnology, PNMs offer versatile biomedical applications, particularly in addressing the intricate challenges of TNBC. The synthesis methods of PNMs, explored within the tumor microenvironment using cellular models, showcase their dynamic nature in cancer treatment. The article anticipates the future of TNBC therapeutics through the optimization of PNMs-based strategies, integrating them into photothermal (PT), photodynamic (PT), and hyperthermia therapy (HTT), drug delivery, and active tumor targeting strategies. Advancements in synthetic methods, coupled with a nuanced understanding of the tumor microenvironment, hold promise for personalized interventions. Comparative investigations of therapeutic models and a thorough exploration of polymeric nanoplatforms toxicological perspectives become imperative for ensuring efficacy and safety. We have explored the interdisciplinary collaboration between nanotechnology, oncology, and molecular biology as pivotal in translating PNMs innovations into tangible benefits for TNBC patients.


Subject(s)
Nanostructures , Polymers , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/therapy , Humans , Nanostructures/chemistry , Nanostructures/administration & dosage , Polymers/chemistry , Female , Animals , Theranostic Nanomedicine/methods , Drug Delivery Systems/methods , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Tumor Microenvironment/drug effects , Hyperthermia, Induced/methods
17.
Int J Pharm ; 660: 124335, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38897488

ABSTRACT

Nanoparticle-mediated thermotherapeutic research strives innovative, multifunctional, efficient, and safe treatments. Our study introduces a novel nanoplatform: the hollow magnetic vortex nanorings within a polydopamine layer (HMVNp), which exhibit dual functionality as magnetic and photothermal agents. Utilizing a "Dual-mode" approach, combining an alternating magnetic field (AMF) with near-infrared (NIR) laser irradiation, HMVNp demonstrated a significant enhancement in heating efficacy (58 ± 8 %, SAR = 1441 vs 1032 W/g) over traditional solid magnetite nanoparticles coated with polydopamine (SMNp). The unique geometry larger surface area to volume ratio facilitates efficient magnetic vortex dynamics and enhanced heat transfer. Addressing the challenge of heat resistant heat shock protein (Hsp) expression, encapsulated quercetin (Q) within HMVNp leverages tumor acidity and dual-mode thermal therapy to enhance release, showing a 28.8 ± 6.81 % increase in Q loading capacity compared to traditional SMNp. Moreover, HMVNp significantly improves contrast for both magnetic resonance imaging (MRI) and photoacoustic imaging (PAI), with an approximately 62 % transverse relaxation (R2 = 81.5 vs 31.6 mM-1s-1 [Fe]). In vivo studies showed that while single treatments slowed tumor growth, dual-mode therapy with quercetin significantly reduced tumors and effectively prevented metastases. Our study highlights the potential of HMVNp/Q as a versatile agent in thermotherapeutic interventions, offering improved diagnostic imaging capabilities.


Subject(s)
Hyperthermia, Induced , Indoles , Magnetic Resonance Imaging , Polymers , Quercetin , Quercetin/administration & dosage , Quercetin/chemistry , Quercetin/pharmacology , Indoles/chemistry , Indoles/administration & dosage , Polymers/chemistry , Animals , Magnetic Resonance Imaging/methods , Hyperthermia, Induced/methods , Mice , Theranostic Nanomedicine/methods , Cell Line, Tumor , Photoacoustic Techniques/methods , Magnetite Nanoparticles/chemistry , Humans , Female , Mice, Nude , Mice, Inbred BALB C , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Photothermal Therapy/methods , Nanoparticles/chemistry
18.
Lancet ; 403(10446): 2759-2761, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38851204
19.
Biomacromolecules ; 25(7): 4233-4245, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38838045

ABSTRACT

In the area of drug delivery aided by stimuli-responsive polymers, the biodegradability of nanocarriers is one of the major challenges that needs to be addressed with the utmost sincerity. Herein, a hydrogen sulfide (H2S) responsive hydrophobic dansyl-based trigger molecule is custom designed and successfully incorporated into the water-soluble polyurethane backbone, which is made of esterase enzyme susceptible urethane bonds. The amphiphilic polyurethanes, PUx (x = 2 and 3) with a biotin chain end, formed self-assembled nanoaggregates. A hemolysis and cytotoxicity profile of doxorubicin (DOX)-loaded biotinylated PU3 nanocarriers revealed that it is nonhemolytic and has excellent selectivity toward HeLa cells (biotin receptor-positive cell lines) causing ∼60% cell death while maintaining almost 100% cell viability for HEK 293T cells (biotin receptor-negative cell lines). Furthermore, better cellular internalization of DOX-loaded fluorescent nanocarriers in HeLa cells than in HEK 293T cells confirmed receptor-mediated endocytosis. Thus, this work ensures that the synthesized polymers serve as biodegradable nanocarriers for anticancer therapeutics.


Subject(s)
Doxorubicin , Drug Delivery Systems , Polyurethanes , Humans , Polyurethanes/chemistry , HeLa Cells , Doxorubicin/pharmacology , Doxorubicin/chemistry , HEK293 Cells , Drug Delivery Systems/methods , Drug Carriers/chemistry , Theranostic Nanomedicine/methods , Biotinylation , Biotin/chemistry , Cell Survival/drug effects , Nanoparticles/chemistry
20.
Theranostics ; 14(8): 3043-3079, 2024.
Article in English | MEDLINE | ID: mdl-38855174

ABSTRACT

In 1853, the perception of prostate cancer (PCa) as a rare ailment prevailed, was described by the eminent Londoner surgeon John Adams. Rapidly forward to 2018, the landscape dramatically altered. Currently, men face a one-in-nine lifetime risk of PCa, accentuated by improved diagnostic methods and an ageing population. With more than three million men in the United States alone grappling with this disease, the overall risk of succumbing to stands at one in 39. The intricate clinical and biological diversity of PCa poses serious challenges in terms of imaging, ongoing monitoring, and disease management. In the field of theranostics, diagnostic and therapeutic approaches that harmoniously merge targeted imaging with treatments are integrated. A pivotal player in this arena is radiotheranostics, employing radionuclides for both imaging and therapy, with prostate-specific membrane antigen (PSMA) at the forefront. Clinical milestones have been reached, including FDA- and/or EMA-approved PSMA-targeted radiodiagnostic agents, such as [18F]DCFPyL (PYLARIFY®, Lantheus Holdings), [18F]rhPSMA-7.3 (POSLUMA®, Blue Earth Diagnostics) and [68Ga]Ga-PSMA-11 (Locametz®, Novartis/ ILLUCCIX®, Telix Pharmaceuticals), as well as PSMA-targeted radiotherapeutic agents, such as [177Lu]Lu-PSMA-617 (Pluvicto®, Novartis). Concurrently, ligand-drug and immune therapies designed to target PSMA are being advanced through rigorous preclinical research and clinical trials. This review delves into the annals of PSMA-targeted radiotheranostics, exploring its historical evolution as a signature molecule in PCa management. We scrutinise its clinical ramifications, acknowledge its limitations, and peer into the avenues that need further exploration. In the crucible of scientific inquiry, we aim to illuminate the path toward a future where the enigma of PCa is deciphered and where its menace is met with precise and effective countermeasures. In the following sections, we discuss the intriguing terrain of PCa radiotheranostics through the lens of PSMA, with the fervent hope of advancing our understanding and enhancing clinical practice.


Subject(s)
Antigens, Surface , Glutamate Carboxypeptidase II , Prostatic Neoplasms , Radiopharmaceuticals , Humans , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/therapy , Glutamate Carboxypeptidase II/metabolism , Male , Antigens, Surface/metabolism , Radiopharmaceuticals/therapeutic use , Nuclear Medicine/methods , Nuclear Medicine/history , Theranostic Nanomedicine/methods , Radioisotopes/therapeutic use , History, 21st Century , History, 20th Century
SELECTION OF CITATIONS
SEARCH DETAIL