Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.432
Filter
1.
Biomaterials ; 312: 122722, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39096841

ABSTRACT

Ferroptosis, a recently identified form of cell death, holds promise for cancer therapy, but concerns persist regarding its uncontrolled actions and potential side effects. Here, we present a semiconducting polymer nanoprodrug (SPNpro) featuring an innovative ferroptosis prodrug (DHU-CBA7) to induce sono-activatable ferroptosis for tumor-specific therapy. DHU-CBA7 prodrug incorporate methylene blue, ferrocene and urea bond, which can selectively and specifically respond to singlet oxygen (1O2) to turn on ferroptosis action via rapidly cleaving the urea bonds. DHU-CBA7 prodrug and a semiconducting polymer are self-assembled with an amphiphilic polymer to construct SPNpro. Ultrasound irradiation of SPNpro leads to the production of 1O2 via sonodynamic therapy (SDT) of the semiconducting polymer, and the generated 1O2 activated DHU-CBA7 prodrug to achieve sono-activatable ferroptosis. Consequently, SPNpro combine SDT with the controlled ferroptosis to effectively cure 4T1 tumors covered by 2-cm tissue with a tumor inhibition efficacy as high as 100 %, and also completely restrain tumor metastases. This study introduces a novel sono-activatable prodrug strategy for regulating ferroptosis, allowing for precise cancer therapy.


Subject(s)
Ferroptosis , Mice, Inbred BALB C , Polymers , Prodrugs , Semiconductors , Ferroptosis/drug effects , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/therapeutic use , Animals , Polymers/chemistry , Female , Cell Line, Tumor , Mice , Ultrasonic Therapy/methods , Nanoparticles/chemistry , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Singlet Oxygen/metabolism
2.
J Am Chem Soc ; 146(32): 22675-22688, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39088029

ABSTRACT

Redox-responsive homodimer prodrug nanoassemblies (RHPNs) have emerged as a significant technology for overcoming chemotherapeutical limitations due to their high drug-loading capacity, low excipient-associated toxicity, and straightforward preparation method. Previous studies indicated that α-position disulfide bond bridged RHPNs exhibited rapid drug release rates but unsatisfactory assembly stability. In contrast, γ-disulfide bond bridged RHPNs showed better assembly stability but low drug release rates. Therefore, designing chemical linkages that ensure both stable assembly and rapid drug release remains challenging. To address this paradox of stable assembly and rapid drug release in RHPNs, we developed carbon-spaced double-disulfide bond (CSDD)-bridged RHPNs (CSDD-RHPNs) with two carbon-spaces. Pilot studies showed that CSDD-RHPNs with two carbon-spaces exhibited enhanced assembly stability, reduction-responsive drug release, and improved selective toxicity compared to α-/γ-position single disulfide bond bridged RHPNs. Based on these findings, CSDD-RHPNs with four and six carbon-spaces were designed to further investigate the properties of CSDD-RHPNs. These CSDD-RHPNs exhibited excellent assembly ability, safety, and prolonged circulation. Particularly, CSDD-RHPNs with two carbon-spaces displayed the best antitumor efficacy on 4T1 and B16-F10 tumor-bearing mice. CSDD chemical linkages offer novel perspectives on the rational design of RHPNs, potentially overcoming the design limitations regarding contradictory assembly ability and drug release rate.


Subject(s)
Carbon , Disulfides , Prodrugs , Disulfides/chemistry , Prodrugs/chemistry , Animals , Mice , Carbon/chemistry , Humans , Drug Liberation , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Cell Line, Tumor , Nanostructures/chemistry , Dimerization , Doxorubicin/chemistry , Doxorubicin/pharmacology
3.
Molecules ; 29(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39124883

ABSTRACT

This review delves into recent advancements in the field of nitro(het)aromatic bioreductive agents tailored for hypoxic environments. These compounds are designed to exploit the low-oxygen conditions typically found in solid tumors, making them promising candidates for targeted cancer therapies. Initially, this review focused on their role as gene-directed enzyme prodrugs, which are inert until activated by specific enzymes within tumor cells. Upon activation, these prodrugs undergo chemical transformations that convert them into potent cytotoxic agents, selectively targeting cancerous tissue while sparing healthy cells. Additionally, this review discusses recent developments in prodrug conjugates containing nitro(het)aromatic moieties, designed to activate under low-oxygen conditions within tumors. This approach enhances their efficacy and specificity in cancer treatment. Furthermore, this review covers innovative research on using nitro(het)aromatic compounds as fluorescent probes for imaging hypoxic tumors. These probes enable non-invasive visualization of low-oxygen regions within tumors, providing valuable insights for the diagnosis, treatment planning, and monitoring of therapeutic responses. We hope this review will inspire researchers to design and synthesize improved compounds for selective cancer treatment and early diagnostics.


Subject(s)
Fluorescent Dyes , Neoplasms , Prodrugs , Tumor Hypoxia , Humans , Prodrugs/chemistry , Prodrugs/pharmacology , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Fluorescent Dyes/chemistry , Animals , Optical Imaging/methods , Nitro Compounds/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use
4.
Molecules ; 29(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39125024

ABSTRACT

Dimeric prodrugs have been investigated intensely as carrier-free drug self-delivery systems (DSDSs) in recent decades, and their stimuli-responsive drug release has usually been controlled by the conjugations between the drug molecules, including the stimuli (pH or redox) and responsive sensitivity. Here, an acid-triggered dimeric prodrug of doxorubicin (DOX) was synthesized by conjugating two DOX molecules with an acid-labile ketal linker. It possessed high drug content near the pure drug, while the premature drug leakage in blood circulation was efficiently suppressed. Furthermore, its aggregation structures were controlled by fabricating nanomedicines via different approaches, such as fast precipitation and slow self-assembly, to regulate the drug release performance. Such findings are expected to enable better anti-tumor efficacy with the desired drug release rate, beyond the molecular structure of the dimeric prodrug.


Subject(s)
Doxorubicin , Drug Delivery Systems , Drug Liberation , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Humans , Hydrogen-Ion Concentration , Drug Carriers/chemistry , Molecular Structure
5.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125821

ABSTRACT

Chlorambucil-platinum(IV) prodrugs exhibit multi-mechanistic chemotherapeutic activity with promising anticancer potential. The platinum(II) precursors of the prodrugs have been previously found to induce changes in the microtubule cytoskeleton, specifically actin and tubulin of HT29 colon cells, while chlorambucil alkylates the DNA. These prodrugs demonstrate significant anticancer activity in 2D cell and 3D spheroid viability assays. A notable production of reactive oxygen species has been observed in HT29 cells 72 h post treatment with prodrugs of this type, while the mitochondrial membrane potential was substantially reduced. The cellular uptake of the chlorambucil-platinum(IV) prodrugs, assessed by ICP-MS, confirmed that active transport was the primary uptake mechanism, with platinum localisation identified primarily in the cytoskeletal fraction. Apoptosis and necrosis were observed at 72 h of treatment as demonstrated by Annexin V-FITC/PI assay using flow cytometry. Immunofluorescence measured via confocal microscopy showed significant changes in actin and tubulin intensity and in architecture. Western blot analysis of intrinsic and extrinsic pathway apoptotic markers, microtubule cytoskeleton markers, cell proliferation markers, as well as autophagy markers were studied post 72 h of treatment. The proteomic profile was also studied with a total of 1859 HT29 proteins quantified by mass spectroscopy, with several dysregulated proteins. Network analysis revealed dysregulation in transcription, MAPK markers, microtubule-associated proteins and mitochondrial transport dysfunction. This study confirms that chlorambucil-platinum(IV) prodrugs are candidates with promising anticancer potential that act as multi-mechanistic chemotherapeutics.


Subject(s)
Antineoplastic Agents , Apoptosis , Chlorambucil , Cisplatin , Colorectal Neoplasms , Drug Resistance, Neoplasm , Prodrugs , Humans , Chlorambucil/pharmacology , Chlorambucil/chemistry , Prodrugs/pharmacology , Prodrugs/chemistry , Drug Resistance, Neoplasm/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Apoptosis/drug effects , Cisplatin/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , HT29 Cells , Membrane Potential, Mitochondrial/drug effects , Platinum/chemistry , Platinum/pharmacology , Reactive Oxygen Species/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Line, Tumor
6.
ACS Nano ; 18(32): 21156-21170, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39088743

ABSTRACT

How to address the resistance of cisplatin (CDDP) has always been a clinical challenge. The resistance mechanism of platinum-based drugs is very complex, including nuclear DNA damage repair, apoptosis escape, and tumor metabolism reprogramming. Tumor cells can switch between mitochondrial oxidative phosphorylation (OXPHOS) and glycolysis and develop resistance to chemotherapy drugs through metabolic variability. In addition, due to the lack of histone protection and a relatively weak damage repair ability, mitochondrial DNA (mtDNA) is more susceptible to damage, which in turn affects mitochondrial OXPHOS and can become a potential target for platinum-based drugs. Therefore, mitochondria, as targets of anticancer drugs, have become a hot topic in tumor resistance research. This study constructed a self-assembled nanotargeted drug delivery system LND-SS-Pt-TPP/HA-CD. ß-Cyclodextrin-grafted hydronic acid (HA-CD)-encapsulated prodrug nanoparticles can target CD44 on the tumor surface and further deliver the prodrug to intracellular mitochondria through a triphenylphosphine group (TPP+). Disulfide bonds can be selectively degraded by glutathione (GSH) in mitochondria, releasing lonidamine (LND) and the cisplatin prodrug (Pt(IV)). Under the action of GSH and ascorbic acid, Pt(IV) is further reduced to cisplatin (Pt(II)). Cisplatin can cause mtDNA damage, induce mitochondrial dysfunction and mitophagy, and then affect mitochondrial OXPHOS. Meanwhile, LND can reduce the hexokinase II (HK II) level, induce destruction of mitochondria, and block energy supply by glycolysis inhibition. Ultimately, this self-assembled nano targeted delivery system can synergistically kill cisplatin-resistant lung cancer cells, which supplies an overcome cisplatin resistance choice via the disrupt mitochondria therapy.


Subject(s)
Antineoplastic Agents , Cisplatin , Drug Resistance, Neoplasm , Lung Neoplasms , Mitochondria , Prodrugs , Cisplatin/pharmacology , Cisplatin/chemistry , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Prodrugs/pharmacology , Prodrugs/chemistry , Nanoparticles/chemistry , Animals , Mice , Drug Delivery Systems , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Cell Line, Tumor , Metabolic Reprogramming
7.
Eur J Med Chem ; 276: 116699, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39089000

ABSTRACT

Drugs administered through conventional formulations are devoid of targeting and often spread to various undesired sites, leading to sub-lethal concentrations at the site of action and the emergence of undesired effects. Hence, therapeutic agents should be delivered in a controlled manner at target sites. Currently, stimuli-based drug delivery systems have demonstrated a remarkable potential for the site-specific delivery of therapeutic moieties. pH is one of the widely exploited stimuli for drug delivery as several pathogenic conditions such as tumor cells, infectious and inflammatory sites are characterized by a low pH environment. This review article aims to demonstrate various strategies employed in the design of acid-sensitive prodrugs, providing an overview of commercially available acid-sensitive prodrugs. Furthermore, we have compiled the progress made for the development of new acid-sensitive prodrugs currently undergoing clinical trials. These prodrugs include albumin-binding prodrugs (Aldoxorubicin and DK049), polymeric micelle (NC-6300), polymer conjugates (ProLindac™), and an immunoconjugate (IMMU-110). The article encompasses a broad spectrum of studies focused on the development of acid-sensitive prodrugs for anticancer, antibacterial, and anti-inflammatory agents. Finally, the challenges associated with the acid-sensitive prodrug strategy are discussed, along with future directions.


Subject(s)
Antineoplastic Agents , Prodrugs , Animals , Humans , Acids/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Delivery Systems , Drug Liberation , Hydrogen-Ion Concentration , Prodrugs/chemistry , Prodrugs/pharmacology
8.
J Nanobiotechnology ; 22(1): 498, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164657

ABSTRACT

Microcarrier is a promising drug delivery system demonstrating significant value in treating cancers. One of the main goals is to devise microcarriers with ingenious structures and functions to achieve better therapeutic efficacy in tumors. Here, inspired by the nucleus-cytoplasm structure of cells and the material exchange reaction between them, we develop a type of biorthogonal compartmental microparticles (BCMs) from microfluidics that can separately load and sequentially release cyclooctene-modified doxorubicin prodrug (TCO-DOX) and tetrazine-modified indocyanine green (Tz-ICG) for tumor therapy. The Tz-ICG works not only as an activator for TCO-DOX but also as a photothermal agent, allowing for the combination of bioorthogonal chemotherapy and photothermal therapy (PTT). Besides, the modification of DOX with cyclooctene significantly decreases the systemic toxicity of DOX. As a result, the developed BCMs demonstrate efficient in vitro tumor cell eradication and exhibit notable tumor growth inhibition with favorable safety. These findings illustrate that the formulated BCMs establish a platform for bioorthogonal prodrug activation and localized delivery, holding significant potential for cancer therapy and related applications.


Subject(s)
Doxorubicin , Drug Delivery Systems , Indocyanine Green , Photothermal Therapy , Prodrugs , Doxorubicin/pharmacology , Doxorubicin/chemistry , Photothermal Therapy/methods , Humans , Prodrugs/pharmacology , Prodrugs/chemistry , Animals , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Mice , Drug Delivery Systems/methods , Cell Line, Tumor , Neoplasms/therapy , Neoplasms/drug therapy , Cyclooctanes/chemistry , Cyclooctanes/pharmacology , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Female
9.
ACS Appl Mater Interfaces ; 16(33): 43272-43282, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39126693

ABSTRACT

Head and neck squamous cell carcinomas are characterized by a high incidence of recurrence, especially in patients with locally advanced disease. Standard treatment strategies can be associated with severe side effects to healthy tissues that can negatively impact the patient's quality of life. Hyperthermia (HT) is a noninvasive treatment modality that has improved the effectiveness of chemotherapy (CT) and/or radiotherapy (RT) for the management of some solid neoplasms. In this context, the association of this approach with rationally designed nanomaterials may further enhance the treatment outcome. In this study, we demonstrate the enhanced effect of neoadjuvant HT in combination with hybrid nanoarchitectures enclosing a cisplatin prodrug (NAs-CisPt) and RT. All the treatments and their combinations have been fully evaluated by employing standardized chorioallantoic membrane tumor models of HPV-negative head and neck carcinoma. An improved tumor-shrinking effect was observed by the administration of the trimodal treatment (HT/NAs-CisPt/RT), which also highlighted a significant increase in apoptosis. Our findings demonstrate that the combination of HT with nanotechnology-based CT and RT in a certain order enhances the in vivo treatment outcome. On a broader basis, this study paves the way for the next exploration of noninvasive treatment approaches for the clinical management of oral cancer based on innovative strategies.


Subject(s)
Chemoradiotherapy , Head and Neck Neoplasms , Hyperthermia, Induced , Nanostructures , Head and Neck Neoplasms/therapy , Head and Neck Neoplasms/pathology , Humans , Hyperthermia, Induced/methods , Animals , Chemoradiotherapy/methods , Nanostructures/chemistry , Nanostructures/therapeutic use , Neoadjuvant Therapy/methods , Cisplatin/therapeutic use , Cell Line, Tumor , Squamous Cell Carcinoma of Head and Neck/therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/therapeutic use
10.
J Mater Chem B ; 12(33): 8067-8075, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39129477

ABSTRACT

Glucose-6-phosphate dehydrogenase (G6PD) is a promising target in cancer therapy. However, poor cellular uptake and off-target toxicity have impeded the clinical translation of a canonical G6PD inhibitor (6-aminonicotinamide/6AN). Here, we report a prodrug strategy to address this issue. The tailored 6AN prodrug contains an azo-bearing protection moiety. The hydrophobic prodrug showed increased cellular uptake than 6AN and was vulnerable to hypoxia, resulting in NAD(P)H quinone dehydrogenase 1 (NQO1)-triggered cleavage of azo bonds. Intriguingly, the prodrug showed configuration-dependent anti-cancer potency. Despite the lower thermodynamic stability, the cis isomer showed enhanced cellular uptake compared to the trans counterpart due to the increased aqueous solubility. Moreover, the boosted potency of the cis isomer compared to the trans isomer arose from the enhancement of NOQ1-catalyzed 6AN release under hypoxia, a hallmark of solid tumors. The discovery of hypoxia-responsive 6AN prodrugs in the current work opens up new avenues for G6PD-targeting cancer medicines.


Subject(s)
6-Aminonicotinamide , Antineoplastic Agents , NADP , Oxidation-Reduction , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Humans , 6-Aminonicotinamide/pharmacology , 6-Aminonicotinamide/chemistry , NADP/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Molecular Structure , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Cell Hypoxia/drug effects , Drug Screening Assays, Antitumor
11.
Nat Commun ; 15(1): 6608, 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39098906

ABSTRACT

The antitumor performance of PROteolysis-TArgeting Chimeras (PROTACs) is limited by its insufficient tumor specificity and poor pharmacokinetics. These disadvantages are further compounded by tumor heterogeneity, especially the presence of cancer stem-like cells, which drive tumor growth and relapse. Herein, we design a region-confined PROTAC nanoplatform that integrates both reactive oxygen species (ROS)-activatable and hypoxia-responsive PROTAC prodrugs for the precise manipulation of bromodomain and extraterminal protein 4 expression and tumor eradication. These PROTAC nanoparticles selectively accumulate within and penetrate deep into tumors via response to matrix metalloproteinase-2. Photoactivity is then reactivated in response to the acidic intracellular milieu and the PROTAC is discharged due to the ROS generated via photodynamic therapy specifically within the normoxic microenvironment. Moreover, the latent hypoxia-responsive PROTAC prodrug is restored in hypoxic cancer stem-like cells overexpressing nitroreductase. Here, we show the ability of region-confined PROTAC nanoplatform to effectively degrade BRD4 in both normoxic and hypoxic environments, markedly hindering tumor progression in breast and head-neck tumor models.


Subject(s)
Cell Cycle Proteins , Nanoparticles , Proteolysis , Transcription Factors , Humans , Proteolysis/drug effects , Animals , Nanoparticles/chemistry , Cell Line, Tumor , Mice , Transcription Factors/metabolism , Female , Cell Cycle Proteins/metabolism , Reactive Oxygen Species/metabolism , Prodrugs/pharmacology , Prodrugs/chemistry , Photochemotherapy/methods , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Mice, Nude , Xenograft Model Antitumor Assays , Tumor Microenvironment/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Nuclear Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Bromodomain Containing Proteins
12.
J Nanobiotechnology ; 22(1): 468, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103846

ABSTRACT

Ulcerative colitis (UC) is a challenging inflammatory gastrointestinal disorder, whose therapies encounter limitations in overcoming insufficient colonic retention and rapid systemic clearance. In this study, we report an innovative polymeric prodrug nanoformulation for targeted UC treatment through sustained 5-aminosalicylic acid (5-ASA) delivery. Amphiphilic polymer-based 13.5 nm micelles were engineered to incorporate azo-linked 5-ASA prodrug motifs, enabling cleavage via colonic azoreductases. In vitro, micelles exhibited excellent stability under gastric/intestinal conditions while demonstrating controlled 5-ASA release over 24 h in colonic fluids. Orally administered micelles revealed prolonged 24-h retention and a high accumulation within inflamed murine colonic tissue. At an approximately 60% dose reduction from those most advanced recent studies, the platform halted DSS colitis progression and outperformed standard 5-ASA therapy through a 77-97% suppression of inflammatory markers. Histological analysis confirmed intact colon morphology and restored barrier protein expression. This integrated prodrug nanoformulation addresses limitations in colon-targeted UC therapy through localized bioactivation and tailored pharmacokinetics, suggesting the potential of nanotechnology-guided precision delivery to transform disease management.


Subject(s)
Colitis , Colon , Delayed-Action Preparations , Mesalamine , Micelles , Nitroreductases , Polymers , Prodrugs , Animals , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Mesalamine/chemistry , Mesalamine/pharmacokinetics , Nitroreductases/metabolism , Mice , Colon/metabolism , Colon/pathology , Polymers/chemistry , Colitis/drug therapy , Colitis/metabolism , Delayed-Action Preparations/chemistry , NADH, NADPH Oxidoreductases/metabolism , Mice, Inbred C57BL , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Male
13.
ACS Nano ; 18(34): 23497-23507, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39146387

ABSTRACT

Colorectal cancer (CRC) is a major global health concern, and the development of effective treatment strategies is crucial. Enzyme prodrug therapy (EPT) shows promise in combating tumors but faces challenges in achieving sustained expression of therapeutic enzymes and optimal biological distribution. To address these issues, a fungi-triggered in situ chemotherapeutics generator (named as SC@CS@5-FC) was constructed via oral delivery of a prodrug (5-fluorocytosine, 5-FC) for the treatment of orthotopic colorectal tumor. When SC@CS@5-FC targets the tumor through tropism by Saccharomyces cerevisiae (SC), the chemotherapeutic generator could be degraded under abundant hyaluronidase (HAase) in the tumor microenvironment by an enzyme-responsive gate to release prodrug (5-FC). And nontoxic 5-FC was catalyzed to toxic chemotherapy drug 5-fluorouracil (5-FU) by cytosine deaminase (CD) of SC. Meanwhile, SC and zinc-coordinated chitosan nanoparticles could be used as immune adjuvants to activate antigen-presenting cells and further enhance the therapeutic effect. Our results demonstrated that SC@CS@5-FC could effectively inhibit tumor growth and prolong mouse survival in an orthotopic colorectal cancer model. This work utilizes living SC as a dynamotor and positioning system for the chemotherapeutic generator SC@CS@5-FC, providing a strategy for oral enzyme prodrug therapy for the treatment of orthotopic colorectal.


Subject(s)
Colorectal Neoplasms , Flucytosine , Fluorouracil , Immunotherapy , Prodrugs , Saccharomyces cerevisiae , Prodrugs/chemistry , Prodrugs/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Animals , Mice , Humans , Flucytosine/pharmacology , Flucytosine/chemistry , Administration, Oral , Fluorouracil/pharmacology , Fluorouracil/chemistry , Fluorouracil/administration & dosage , Cytosine Deaminase/metabolism , Chitosan/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Hyaluronoglucosaminidase/metabolism , Mice, Inbred BALB C , Nanoparticles/chemistry , Drug Screening Assays, Antitumor
14.
Carbohydr Polym ; 343: 122489, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39174141

ABSTRACT

The clinical utility of chemotherapy is often compromised by its limited efficacy and significant side effects. Addressing these concerns, we have developed a self-assembled nanomicelle, namely SANTA FE OXA, which consists of hyaluronic acid (HA) conjugated with ferrocene methanol (FC), oxaliplatin prodrug (OXA(IV)) and ethylene glycol-coupled linoleic acid (EG-LA). Targeted delivery is achieved by HA binding to the CD44 receptors that are overexpressed on tumor cells, facilitating drug uptake. Once internalized, hyaluronidase (HAase) catalyzes the digestion of the SANTA FE OXA, releasing FC and reducing OXA(IV) into an active form. The active oxaliplatin (OXA) induces DNA damage and increases intracellular hydrogen peroxide (H2O2) levels via cascade reactions. Simultaneously, FC disrupts the redox balance within tumor cells, inducing ferroptosis. Both in vivo and in vitro experiments confirmed that SANTA FE OXA inhibited tumor growth by combining cascade chemotherapy and self-sensitized ferroptosis, achieving a tumor inhibition rate of up to 76.61 %. Moreover, this SANTA FE OXA significantly mitigates the systemic toxicity commonly associated with platinum-based chemotherapeutics. Our findings represent a compelling advancement in nanomedicine for enhanced cascade cancer therapy.


Subject(s)
Antineoplastic Agents , Ferroptosis , Ferrous Compounds , Hyaluronic Acid , Micelles , Oxaliplatin , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Ferroptosis/drug effects , Oxaliplatin/pharmacology , Oxaliplatin/chemistry , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice , Cell Line, Tumor , Ferrous Compounds/chemistry , Ferrous Compounds/pharmacology , Metallocenes/chemistry , Metallocenes/pharmacology , Prodrugs/pharmacology , Prodrugs/chemistry , Linoleic Acid/chemistry , Linoleic Acid/pharmacology , Mice, Inbred BALB C , Female , Mice, Nude , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/pharmacology , Neoplasms/drug therapy
15.
Biomacromolecules ; 25(9): 5834-5846, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39191734

ABSTRACT

Bioorthogonal reactions present a promising strategy for minimizing off-target toxicity in cancer chemotherapy, yet a dependable nanoplatform is urgently required. Here, we have fabricated an acid-responsive polymer micelle for the specific delivery and activation of the prodrug within tumor cells through Ru catalyst-mediated bioorthogonal reactions. The decomposition of micelles, triggered by the cleavage of the hydrazone bond in the acidic lysosomal environment, facilitated the concurrent release of Alloc-DOX and the Ru catalyst within the cells. Subsequently, the uncaging process of Alloc-DOX was demonstrated to be induced by the high levels of glutathione within tumor cells. Notably, the limited glutathione inside normal cells prevented the conversion of Alloc-DOX into active DOX, thereby minimizing the toxicity toward normal cells. In tumor-bearing mice, this nanoplatform exhibited enhanced efficacy in tumor suppression while minimizing off-target toxicity. Our study provides an innovative approach for in situ drug activation that combines safety and effectiveness in cancer chemotherapy.


Subject(s)
Doxorubicin , Micelles , Polymers , Prodrugs , Ruthenium , Prodrugs/chemistry , Prodrugs/pharmacology , Animals , Humans , Mice , Doxorubicin/pharmacology , Doxorubicin/chemistry , Ruthenium/chemistry , Polymers/chemistry , Catalysis , Drug Delivery Systems/methods , Cell Line, Tumor , Mice, Inbred BALB C , Mice, Nude
16.
Molecules ; 29(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39202916

ABSTRACT

The diselenide bond has attracted intense interest for drug delivery systems (DDSs) for tumor chemotherapy, owing to it possessing higher redox sensitivity than the disulfide one. Various redox-responsive diselenide-containing carriers have been developed for chemotherapeutics delivery. However, the premature drug leakage from these DDSs was significant enough to cause toxic side effects on normal cells. Here, a pH/redox co-triggered degradable polyprodrug was designed as a drug self-delivery system (DSDS) by incorporating drug molecules as structural units in the polymer main chains, using a facile one-pot two-step approach. The proposed PDOX could only degrade and release drugs by breaking both the neighboring acid-labile acylhydrazone and the redox-cleavable diselenide conjugations in the drug's structural units, triggered by the higher acidity and glutathione (GSH) or reactive oxygen species (ROS) levels in the tumor cells. Therefore, a slow solubility-controlled drug release was achieved for tumor-specific chemotherapy, indicating promising potential as a safe and efficient long-acting DSDS for future tumor treatment.


Subject(s)
Antineoplastic Agents , Oxidation-Reduction , Prodrugs , Hydrogen-Ion Concentration , Humans , Prodrugs/chemistry , Prodrugs/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Liberation , Reactive Oxygen Species/metabolism , Drug Delivery Systems , Drug Carriers/chemistry , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/metabolism , Polymers/chemistry , Glutathione/chemistry , Glutathione/metabolism , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/administration & dosage
17.
Chem Commun (Camb) ; 60(71): 9566-9569, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39139058

ABSTRACT

Herein, we report the first-ever design strategy of modifying RAPTA-C into a self-reporting prodrug candidate based on Ru-coordinated polydiacetylene self-assembly. This nanosystem exhibits a dual lock strategy that responds to visible light and pH-stimuli sequentially one by one with a concomitant color change for controlled RAPTA-C release and real-time release monitoring in human gastric cancer cells.


Subject(s)
Polyacetylene Polymer , Polyynes , Prodrugs , Prodrugs/chemistry , Humans , Polyacetylene Polymer/chemistry , Polyynes/chemistry , Hydrogen-Ion Concentration , Drug Liberation , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ruthenium/chemistry , Light , Stomach Neoplasms/drug therapy , Polymers/chemistry , Molecular Structure
18.
Nat Commun ; 15(1): 7558, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214974

ABSTRACT

Hydrogen sulfide is essential in numerous physiological and pathological processes and has emerged as a promising cancer imaging and signaling molecule and a potentially versatile therapeutic agent. However, the endogenous levels of hydrogen sulfide remain insufficient to perform its biological functions, and thus, developing novel strategies that amplify hydrogen sulfide signals at lesion sites is of increasing interest. In this work, a nanoplatform (SNP) based on hydrogen sulfide-responsive self-immolative poly(thiocarbamate) with localized hydrogen sulfide signal amplification capability is developed to encapsulate a hydrogen sulfide-responsive fluorescent probe (e.g., hemicyanine dye; p-Cy) or an anticancer prodrug (e.g., doxorubicin; p-DOX) to form a nanoprobe (SNPp-Cy) or nanomedicine (SNPp-DOX) for cancer imaging and therapy, respectively. SNPp-Cy exhibits a low detection limit for hydrogen sulfide, enabling ultrasensitive detection of small (<2 mm) tumors in female mice. In addition, SNPp-DOX can effectively inhibit the growth of DOX-resistant human breast cancer xenograft, lung metastasis, and patient-derived xenograft tumors in female mice.


Subject(s)
Doxorubicin , Hydrogen Sulfide , Hydrogen Sulfide/metabolism , Animals , Humans , Female , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Doxorubicin/administration & dosage , Mice , Cell Line, Tumor , Fluorescent Dyes/chemistry , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Mice, Nude , Xenograft Model Antitumor Assays , Prodrugs/pharmacology , Prodrugs/therapeutic use , Prodrugs/chemistry , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Mice, Inbred BALB C , Nanoparticles/chemistry , Optical Imaging/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
19.
Chem Commun (Camb) ; 60(67): 8892-8895, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39086281

ABSTRACT

A biological nanoplatform (Gal-ANI@ZnAP NPs) was constructed based on a prodrug-skeletal metal-organic framework (MOF) using purine nucleobase analogue prodrug 6-allylthiopurine as a bioactive ligand, and functionalized with AIE fluorescent PARP inhibitor glycoconjugate for visualization therapy and synthetic lethal cancer therapy. This nanoplatform could actively target cancer cells, selectively release drugs in response to esterase/pH, and visualize drug uptake. In vitro studies revealed that Gal-ANI@ZnAP NPs increased the synthetic lethality in cancer cells by inducing DNA repair failure with the simultaneous targeting of PARP and nucleotide metabolism, thereby exhibiting a significant cancer-killing effect. The study presents a novel strategy to construct an AIE nanoplatform using pharmaceutical molecules for drug uptake visualization and boosting synthetic lethality in cancer.


Subject(s)
Antineoplastic Agents , Metal-Organic Frameworks , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Glycosylation , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Cell Line, Tumor , Nanoparticles/chemistry , Drug Screening Assays, Antitumor , Cell Survival/drug effects
20.
J Nanobiotechnology ; 22(1): 485, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138462

ABSTRACT

Nanozymes are promising antimicrobials, as they produce reactive oxygen species (ROS). However, the intrinsic lack of selectivity of ROS in distinguishing normal flora from pathogenic bacteria deprives nanozymes of the necessary selectivities of ideal antimicrobials. Herein, we exploit the physiological conditions of bacteria (high alkaline phosphatase (ALP) expression) using a novel CuO nanoparticle (NP) nanoenzyme system to initiate an ALP-activated ROS prodrug system for use in the on-demand precision killing of bacteria. The prodrug strategy involves using 2-phospho-L-ascorbic acid trisodium salt (AAP) that catalyzes the ALP in pathogenic bacteria to generate ascorbic acid (AA), which is converted by the CuO NPs, with intrinsic ascorbate oxidase- and peroxidase-like activities, to produce ROS. Notably, the prodrug system selectively kills Escherichia coli (pathogenic bacteria), with minimal influence on Staphylococcus hominis (non-pathogenic bacteria) due to their different levels of ALP expression. Compared to the CuO NPs/AA system, which generally depletes ROS during storage, CuO NPs/AAP exhibits a significantly higher stability without affecting its antibacterial activity. Furthermore, a rat model is used to indicate the applicability of the CuO NPs/AAP fibrin gel in wound disinfection in vivo with negligible side effects. This study reveals the therapeutic precision of this bifunctional tandem nanozyme platform against pathogenic bacteria in ALP-activated conditions.


Subject(s)
Alkaline Phosphatase , Anti-Bacterial Agents , Copper , Disinfection , Escherichia coli , Prodrugs , Reactive Oxygen Species , Copper/chemistry , Copper/pharmacology , Animals , Prodrugs/pharmacology , Prodrugs/chemistry , Alkaline Phosphatase/metabolism , Rats , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Reactive Oxygen Species/metabolism , Disinfection/methods , Ascorbic Acid/pharmacology , Ascorbic Acid/chemistry , Ascorbic Acid/analogs & derivatives , Metal Nanoparticles/chemistry , Rats, Sprague-Dawley , Male
SELECTION OF CITATIONS
SEARCH DETAIL