Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.428
Filter
1.
Drug Dev Res ; 85(6): e22258, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39253992

ABSTRACT

The 2-nitroimidazole based 99mTc-radiopharmaceuticals are widely explored for imaging tumor hypoxia. Radiopharmaceuticals for targeting hypoxia are often lipophilic and therefore, show significant uptake in liver and other vital organs. In this context, lipophilic radiopharmaceuticals with design features enabling faster clearance from liver may be more desirable. A dipicolylamine-NCS bifunctional chelator that could generate a thiourea-bridge up on conjugation to primary amine bearing molecule was used to synthesize a 2-nitroimidazole-dipicolyl amine ligand for radiolabeling with 99mTc(CO)3 core. Corresponding Re(CO)3-analogue was prepared to establish the structure of 2-nitroimidazole-99mTc(CO)3 complex prepared in trace level. The 2-nitroimidazole-99mTc(CO)3 complex showed a hypoxic to normoxic ratio of ~2.5 in CHO cells at 3 h. In vivo, the complex showed accumulation and retention in tumor with high tumor to blood and tumor to muscle ratio. The study demonstrated the utility of metabolizable thiourea-bridge in 2-nitroimidazole-99mTc(CO)3 complex in inducing faster clearance of the radiotracer from liver. The dipicolylamine-NCS bifunctional chelator reported herein can also be used for radiolabeling other class of target specific molecules with 99mTc(CO)3 core.


Subject(s)
Radiopharmaceuticals , Thiourea , Tumor Hypoxia , Animals , Radiopharmaceuticals/pharmacokinetics , CHO Cells , Thiourea/analogs & derivatives , Thiourea/pharmacokinetics , Thiourea/chemistry , Cricetulus , Mice , Nitroimidazoles/pharmacokinetics , Nitroimidazoles/chemistry , Organotechnetium Compounds/pharmacokinetics , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/administration & dosage , Tissue Distribution , Picolinic Acids/pharmacokinetics , Picolinic Acids/chemistry , Humans , Technetium
2.
ACS Nano ; 18(33): 22153-22171, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39118372

ABSTRACT

cGAS/STING pathway, which is highly related to tumor hypoxia, is considered as a potential target for remodeling the immunosuppressive microenvironment of solid tumors. Metal ions, such as Mn2+, activate the cGAS/STING pathway, but their efficacy in cancer therapy is limited by insufficient effect on immunogenic tumor cell death of a single ion. Here, we evaluate the association between tumor hypoxia and cGAS/STING inhibition and report a polymetallic-immunotherapy strategy based on large mesoporous trimetal-based nanozyme (AuPdRh) coordinated with Mn2+ (Mn2+@AuPdRh) to activate cGAS/STING signaling for robust adaptive antitumor immunity. Specifically, the inherent CAT-like activity of this polymetallic Mn2+@AuPdRh nanozyme decomposes the endogenous H2O2 into O2 to relieve tumor hypoxia induced suppression of cGAS/STING signaling. Moreover, the Mn2+@AuPdRh nanozyme displays a potent near-infrared-II photothermal effect and strong POD-mimic activity; and the generated hyperthermia and •OH radicals synergistically trigger immunogenic cell death in tumors, releasing abundant dsDNA, while the delivered Mn2+ augments the sensitivity of cGAS to dsDNA and activates the cGAS-STING pathway, thereby triggering downstream immunostimulatory signals to kill primary and distant metastatic tumors. Our study demonstrates the potential of metal-based nanozyme for STING-mediated tumor polymetallic-immunotherapy and may inspire the development of more effective strategies for cancer immunotherapy.


Subject(s)
Immunotherapy , Infrared Rays , Membrane Proteins , Animals , Mice , Membrane Proteins/metabolism , Manganese/chemistry , Manganese/pharmacology , Nucleotidyltransferases/metabolism , Porosity , Signal Transduction/drug effects , Humans , Tumor Hypoxia/drug effects , Gold/chemistry , Gold/pharmacology , Cell Line, Tumor , Palladium/chemistry , Palladium/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Female
3.
Int J Nanomedicine ; 19: 8211-8236, 2024.
Article in English | MEDLINE | ID: mdl-39157736

ABSTRACT

Hypoxia, as a prominent feature of the tumor microenvironment, has a profound impact on the multicomponent changes within this environment. Under hypoxic conditions, the malignant phenotype of tumor cells, the variety of cell types within the tumor microenvironment, as well as intercellular communication and material exchange, undergo complex alterations. These changes provide significant prospects for exploring the mechanisms of tumor development under different microenvironmental conditions and for devising therapeutic strategies. Exosomes secreted by tumor cells and stromal cells are integral components of the tumor microenvironment, serving as crucial mediators of intercellular communication and material exchange, and have consequently garnered increasing attention from researchers. This review focuses on the mechanisms by which hypoxic conditions promote the release of exosomes by tumor cells and alter their encapsulated contents. It also examines the effects of exosomes derived from tumor cells, immune cells, and other cell types under hypoxic conditions on the tumor microenvironment. Additionally, we summarize current research progress on the potential clinical applications of exosomes under hypoxic conditions and propose future research directions in this field.


Subject(s)
Cell Communication , Exosomes , Neoplasms , Tumor Microenvironment , Exosomes/metabolism , Humans , Cell Communication/physiology , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cell Hypoxia/physiology , Tumor Hypoxia , Hypoxia/metabolism
4.
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
5.
Int J Pharm ; 663: 124581, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39137819

ABSTRACT

Cold tumors lack T cells infiltration and have low immunogenicity, resulting insufficient immunotherapy response. Therefore, how to realize the transformation from cold tumor to hot tumor is an urgent problem to be solved. Photodynamic therapy can induce immunogenic death of tumor cells (ICD) and activate T lymphocytes to produce tumor immune response. However, hypoxia in the cold tumor microenvironment limits the effectiveness of photodynamic therapy. So in this article, MET-HMME/CAT-HMME@Nlip as a functional co-delivery nanoliposomes was constructed based on overcoming the above problems. Firstly, the oxygen-deficient state could be improved by the following two ways, one is catalase loaded in CAT-HMME@Nlip can decompose high concentration hydrogen peroxide to produce oxygen, and the other is metformin loaded in MET-HMME@Nlip can decrease oxygen consumption by inhibiting of mitochondrial respiration. And then with the increase of substrate oxygen concentration, the sensitivity of photodynamic therapy can be greatly improved and the anti-tumor immune response by PDT-induced ICD can also be enhanced obviously. In addition, metformin could act as a small molecule immune checkpoint inhibitor to reduce the expression of PD-L1 on the surface of tumor cells, thereby effectively improving the specific killing ability of cytotoxic T cells to tumor cells which could not only erasing the primary tumor, but also inhibiting the growth of simulated distant tumors through the immune memory function. This study provides a new idea for improving the clinical treatment effect of hypoxic cold tumors, especially for tumors that could not benefit from immunotherapy due to low or no expression of PD-L1 protein on the surface of tumor cells.


Subject(s)
B7-H1 Antigen , Immunotherapy , Liposomes , Metformin , Nanoparticles , Photochemotherapy , Tumor Microenvironment , Photochemotherapy/methods , Animals , Metformin/administration & dosage , Metformin/pharmacology , Immunotherapy/methods , Tumor Microenvironment/drug effects , Cell Line, Tumor , Nanoparticles/administration & dosage , Mice , Catalase/administration & dosage , Humans , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/immunology , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/pharmacology , Female , Photosensitizing Agents/administration & dosage , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Hypoxia/drug effects , Hydrogen Peroxide , Mice, Inbred BALB C
6.
Inorg Chem ; 63(35): 16404-16417, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39150967

ABSTRACT

The limited optical penetration depth and hypoxic tumor microenvironment (TME) are key factors that hinder the practical applications of conventional photodynamic therapy (PDT). To fundamentally address these issues, self-luminescent photosensitizers (PSs) can achieve efficient PDT. Herein, a self-chemiluminescence (CL)-triggered Ir complex PS, namely, IrL2, with low-O2-dependence type I photochemical processes is reported for efficient PDT. The rational design achieves efficient chemiluminescence resonance energy transfer (CRET) from covalently bonded luminol units to the Ir complex in IrL2 under the catalysis of H2O2 and hemoglobin (Hb) to generate O2•- and 1O2. Liposome IrL2H nanoparticles (NPs) are constructed by loading IrL2 and Hb. The intracellular H2O2 and loaded Hb catalyze the luminol part of IrL2H, and the Ir2 part is then excited to produce types I and II reactive oxygen species (ROS) through CRET, inducing cell death, even under hypoxic conditions, and promoting cell apoptosis. IrL2H is used for tumor imaging and inhibits tumor growth in 4T1-bearing mouse models through intratumoral injection without external light sources. This work provides new designs for transition metal complex PSs that conquer the limitations of external light sources and the hypoxic TME in PDT.


Subject(s)
Iridium , Photochemotherapy , Photosensitizing Agents , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Animals , Iridium/chemistry , Iridium/pharmacology , Mice , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Luminescence , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Humans , Mice, Inbred BALB C , Cell Line, Tumor , Drug Screening Assays, Antitumor , Tumor Hypoxia/drug effects , Cell Proliferation/drug effects , Female , Cell Survival/drug effects , Reactive Oxygen Species/metabolism , Molecular Structure
7.
Sci Rep ; 14(1): 20157, 2024 08 30.
Article in English | MEDLINE | ID: mdl-39215035

ABSTRACT

To evaluate the prognostic significance of the maximum standardized uptake value (SUVmax) in nasopharyngeal carcinoma (NPC), establish a gene signature that correlates with SUVmax, and explore the underlying biological behaviors associated with these correlations for the prediction of clinical outcomes. A cohort of 726 patients with NPC was examined to identify correlations between SUVmax and various clinical variables. RNA sequencing was performed to identify genes related to SUVmax, and these genes were used to develop an SUV signature. Additionally, transcriptome enrichment analysis was conducted to investigate the potential biological behaviors underlying the observed correlations. Higher SUVmax was associated with an increased tumor burden and worse prognosis. The SUV signature, which consisted of 10 genes, was positively correlated with SUVmax, and it predicted worse survival outcomes. This signature was highly expressed in malignant epithelial cells and associated with hypoxia and resistance to radiotherapy. Additionally, the signature was negatively correlated with immune function. SUVmax is a valuable prognostic indicator in NPC, with higher values predicting worse outcomes. The SUV signature offers further prognostic insights, linking glucose metabolism to tumor aggressiveness, treatment resistance, and immune function, and it could represent a potential biomarker for NPC.


Subject(s)
Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Tumor Hypoxia , Humans , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/mortality , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/mortality , Male , Female , Tumor Hypoxia/genetics , Prognosis , Middle Aged , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Adult , Transcriptome , Aged , Gene Expression Profiling
8.
J Mol Neurosci ; 74(3): 63, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967861

ABSTRACT

High-grade gliomas (HGG) comprising WHO grades 3 and 4 have a poor overall survival (OS) that has not improved in the past decade. Herein, markers representing four components of the tumor microenvironment (TME) were identified to define their linked expression in TME and predict the prognosis in HGG, namely, interleukin6 (IL6, inflammation), inducible nitric oxide synthase(iNOS), heat shock protein-70 (HSP70, hypoxia), vascular endothelial growth receptor (VEGF), and endothelin1 (ET1) (angiogenesis) and matrix metalloprotease-14 (MMP14) and intercellular adhesion molecule1 (ICAM1, extracellular matrix). To establish a non-invasive panel of biomarkers for precise prognostication in HGG. Eighty-six therapy-naive HGG patients with 45 controls were analyzed for the defined panel. Systemic expression of extracellular/secretory biomarkers was screened dot-immune assay (DIA), quantified by ELISA, and validated by immunocytochemistry (ICC). Expression of iNOS, HSP70, IL-6, VEGF, ET1, MMP14, and ICAM1 was found to be positively associated with grade. Quantification of circulating levels of the markers by ELISA and ICC presented a similar result. The biomarkers were observed to negatively correlate with OS (p < 0.0001). Cox-regression analysis yielded all biomarkers as good prognostic indicators and independent of confounders. On applying combination statistics, the biomarker panel achieved higher sensitivity than single markers to define survival. The intra-association of all seven biomarkers was significant, hinting of a cross-talk between the TME components and a hypoxia driven systemic inflammation upregulating the expression of other components. This is a first ever experimental study of a marker panel that can distinguish between histopathological grades and also delineate differential survival using liquid biopsy, suggesting that markers of hypoxia can be a cornerstone for personalized therapy. The panel of biomarkers of iNOS, HSP70, IL-6, VEGF, ET1, MMP14, and ICAM1 holds promise for prognostication in HGG.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Glioma , HSP70 Heat-Shock Proteins , Neovascularization, Pathologic , Nitric Oxide Synthase Type II , Tumor Microenvironment , Humans , Glioma/metabolism , Glioma/pathology , Female , Male , Middle Aged , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/blood , Biomarkers, Tumor/metabolism , Nitric Oxide Synthase Type II/metabolism , Adult , Neovascularization, Pathologic/metabolism , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/blood , Interleukin-6/metabolism , Interleukin-6/blood , Matrix Metalloproteinase 14/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/blood , Endothelin-1/metabolism , Endothelin-1/blood , Aged , Tumor Hypoxia , Prognosis , Angiogenesis
9.
Int J Pharm ; 662: 124453, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39013531

ABSTRACT

Nanozymes, nanostructured materials emulating natural enzyme activities, exhibit potential in catalyzing reactive oxygen species (ROS) production for cancer treatment. By facilitating oxidative reactions, elevating ROS levels, and influencing the tumor microenvironment (TME), nanozymes foster the eradication of cancer cells. Noteworthy are their superior stability, ease of preservation, and cost-effectiveness compared to natural enzymes, rendering them invaluable for medical applications. This comprehensive review intricately explores the interplay between ROS and tumor therapy, with a focused examination of metal-based nanozyme strategies mitigating tumor hypoxia. It provides nuanced insights into diverse catalytic processes, mechanisms, and surface modifications of various metal nanozymes, shedding light on their role in intra-tumoral ROS generation and applications in antioxidant therapy. The review concludes by delineating specific potential prospects and challenges associated with the burgeoning use of metal nanozymes in future tumor therapies.


Subject(s)
Neoplasms , Reactive Oxygen Species , Tumor Microenvironment , Humans , Reactive Oxygen Species/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Tumor Microenvironment/drug effects , Nanostructures/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Metals/chemistry , Tumor Hypoxia/drug effects
10.
Magy Onkol ; 68(2): 126-135, 2024 Jul 16.
Article in Hungarian | MEDLINE | ID: mdl-39013086

ABSTRACT

Tumor hypoxia plays an important role in controlling tumor progression through signaling pathways related to the transcription factor HIF-1. In addition to enhancing migration, promoting angiogenesis and regulating metabolism, the hypoxic environment also affects immune function. In this hypoxic microenvironment an immunosuppressive milieu is established, where HIF-1 upregulates the expression of PD-L1, a key regulator of the immune response. We have found that elevated expression of PD-L1 correlates with increased HIF-1 levels in cancer cell lines and clinical samples. Thus, the co-inhibition of HIF-1 and PD-1/PD-L1 offers promising therapeutic possibilities. In this review we have examined the limitations of HIF-1 and PD-1/PD-L1 inhibition as monotherapy, explored their combined benefits and evaluated the feasibility of targeting PD-L1 with HIF-1 inhibitors.


Subject(s)
B7-H1 Antigen , Neoplasms , Programmed Cell Death 1 Receptor , Tumor Hypoxia , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Hypoxia/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
11.
Cancer Res Commun ; 4(8): 2008-2024, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39007350

ABSTRACT

Treatment of patients with locally advanced rectal cancer (RC) is based on neoadjuvant chemoradiotherapy followed by surgery. In order to reduce the development of therapy resistance, it is necessary to further improve previous treatment approaches. Recent in vivo experimental studies suggested that the reduction of tumor hypoxia by tumor vessel normalization (TVN), through the inhibition of the glycolytic activator PFKFB3, could significantly improve tumor response to therapy. We have evaluated in vitro and in vivo the effects of the PFKFB3 inhibitor 2E-3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) on cell survival, clonogenicity, migration, invasion, and metabolism using colorectal cancer cells, patient-derived tumor organoid (PDO), and xenograft (PDX). 3PO treatment of colorectal cancer cells increased radiation-induced cell death and reduced cancer cell invasion. Moreover, gene set enrichment analysis shows that 3PO is able to alter the metabolic status of PDOs toward oxidative phosphorylation. Additionally, in vivo neoadjuvant treatment with 3PO induced TVN, alleviated tumor hypoxia, and increased tumor necrosis. Our results support PFKFB3 inhibition as a possible future neoadjuvant addition for patients with RC. SIGNIFICANCE: Novel therapies to better treat colorectal cancer are necessary to improve patient outcomes. Therefore, in this study, we evaluated the combination of a metabolic inhibitor (3PO) and standard radiotherapy in different experimental settings. We have observed that the addition of 3PO increased radiation effects, ultimately improving tumor cell response to therapy.


Subject(s)
Phosphofructokinase-2 , Rectal Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Necrosis , Neoadjuvant Therapy/methods , Neovascularization, Pathologic/drug therapy , Phosphofructokinase-2/antagonists & inhibitors , Pyridines/pharmacology , Pyridines/therapeutic use , Rectal Neoplasms/drug therapy , Rectal Neoplasms/radiotherapy , Tumor Hypoxia/drug effects , Xenograft Model Antitumor Assays
12.
Biomater Sci ; 12(16): 4083-4102, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39010783

ABSTRACT

Hydrogen peroxide (H2O2) as a reactive oxygen species produced by cellular metabolism can be used in antitumor therapy. However, the concentration of intracellular H2O2 limits its application. Some materials could enhance the concentration of intracellular H2O2 to strengthen antitumor therapy. In this review, the recent advances in H2O2-supplying materials in terms of promoting intracellular H2O2 production and exogenous H2O2 supply are summarized. Then the mechanism of H2O2-supplying materials for tumor therapy is discussed from three aspects: reconstruction of the tumor hypoxia microenvironment, enhancement of oxidative stress, and the intrinsic anti-tumor ability of H2O2-supplying materials. In addition, the application of H2O2-supplying materials for tumor therapy is discussed. Finally, the future of H2O2-supplying materials is presented. This review aims to provide a novel idea for the application of H2O2-supplying materials in tumor therapy.


Subject(s)
Hydrogen Peroxide , Neoplasms , Tumor Microenvironment , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Microenvironment/drug effects , Oxidative Stress/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Reactive Oxygen Species/metabolism , Tumor Hypoxia/drug effects
13.
Biomater Adv ; 163: 213962, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39032435

ABSTRACT

Solid tumors create a hypoxic microenvironment and this character can be utilized for cancer therapy, but the hypoxia levels are insufficient to achieve satisfactory therapeutic benefits. Some tactics have been used to improve hypoxia, which however will cause side effects due to the uncontrolled drug release. We herein report near-infrared (NIR) photoactivatable three-in-one nanoagents (PCT) to aggravate tumor hypoxia and enable amplified photo-combinational chemotherapy. PCT are formed based on a thermal-responsive liposome nanoparticle containing three therapeutic agents: a hypoxia responsive prodrug tirapazamine (TPZ) for chemotherapy, a vascular targeting agent combretastatin A-4 (CA4) for vascular disturbance and a semiconducting polymer for both photodynamic therapy (PDT) and photothermal therapy (PTT). With NIR laser irradiation, PCT generate heat for PTT and destructing thermal-responsive liposomes to achieve activatable releases of TPZ and CA4. Moreover, PCT produce singlet oxygen (1O2) for PDT via consuming tumor oxygen. CA4 can disturb the blood vessels in tumor microenvironment to aggravate the hypoxic microenvironment, which results in the activation of TPZ for amplified chemotherapy. PCT thus enable PTT, PDT and hypoxia-amplified chemotherapy to afford a high therapeutic efficacy to almost absolutely eradicate subcutaneous 4 T1 tumors and effectively inhibit tumor metastases in lung and liver. This work presents an activatable three-in-one therapeutic nanoplatform with remotely controllable and efficient therapeutic actions to treat cancer.


Subject(s)
Infrared Rays , Liposomes , Nanoparticles , Photochemotherapy , Tirapazamine , Animals , Humans , Photochemotherapy/methods , Tirapazamine/pharmacology , Tirapazamine/chemistry , Tirapazamine/therapeutic use , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Mice , Tumor Microenvironment/drug effects , Cell Line, Tumor , Photothermal Therapy/methods , Stilbenes/pharmacology , Stilbenes/therapeutic use , Stilbenes/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/therapeutic use , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Tumor Hypoxia/drug effects
14.
ACS Appl Mater Interfaces ; 16(31): 40428-40443, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39042585

ABSTRACT

The spin-orbit charge transfer intersystem crossing (SOCT-ISC) photophysical process has shown great potential for constructing heavy-atom-free photosensitizers (PSs) for photodynamic therapy (PDT) of tumors. However, for almost all such PSs reported to date, the SOCT-ISC is driven by the acceptor-excited photoinduced electron transfer (a-PeT). In this work, for the first time the donor-excited photoinduced electron transfer (d-PeT)-driven SOCT-ISC mechanism is utilized to construct the heavy-atom-free PSs for PDT of tumors by directly installing the electron-deficient N-alkylquinolinium unit (as an electron acceptor) into the meso-position of the near-infrared (NIR) distyryl Bodipy chromophore (as an electron donor). In the less polar environment, the PSs exist as the monomer and promote the production of singlet oxygen (1O2) (Type-II) relying on the d-PeT-driven population of the triplet excited state via SOCT-ISC, whereas in the aqueous environment, they exist as nanoaggregates and induce the generation of superoxides (O2-•) and hydroxyl radicals (HO•) (Type-I) via the d-PeT-driven formation of the delocalized charge-separated state. The PSs could rapidly be internalized into cancer cells and induce the simultaneous production of intracellular 1O2, O2-•, and HO• upon NIR light irradiation, endowing the PSs with superb photocytotoxicity with IC50 values up to submicromolar levels whether under normoxia or under hypoxia. Based on the PSs platform, a tumor-targetable PS is developed, and its abilities in killing cancer cells and in ablating tumors without damage to normal cells/tissues under NIR light irradiation are verified in vitro and in vivo. The study expands the design scope of PSs by introducing the d-PeT conception, thus being highly valuable for achieving novel PSs in the realm of tumor PDT.


Subject(s)
Photochemotherapy , Photosensitizing Agents , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/radiation effects , Humans , Mice , Animals , Electron Transport/drug effects , Singlet Oxygen/metabolism , Singlet Oxygen/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Boron Compounds/chemistry , Boron Compounds/pharmacology , Boron Compounds/radiation effects , Cell Line, Tumor , Infrared Rays , Cell Survival/drug effects , Tumor Hypoxia/drug effects , Mice, Inbred BALB C
15.
ACS Appl Mater Interfaces ; 16(31): 40641-40652, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39051862

ABSTRACT

Photothermal therapy (PTT) has emerged as a noninvasive and precise cancer treatment modality known for its high selectivity and lack of drug resistance. However, the clinical translation of many PTT agents is hindered by the limited biodegradability of inorganic nanoparticles and the instability of organic dyes. In this study, a peptide conjugate, IR820-Cys-Trp-Glu-Trp-Thr-Trp-Tyr (IR820-C), was designed to self-assemble into nanoparticles for both potent PTT and vascular disruption in melanoma treatment. When co-assembled with the poorly soluble vascular disrupting agent (VDA) combretastatin A4 (CA4), the resulting nanoparticles (IR820-C@CA4 NPs) accumulate efficiently in tumors, activate systemic antitumor immune responses, and effectively ablate melanoma with a single treatment and near-infrared irradiation, as confirmed by our in vivo experiments. Furthermore, by exploiting the resulting tumor hypoxia, we subsequently administered the hypoxia-activated prodrug tirapazamine (TPZ) to capitalize on the created microenvironment, thereby boosting therapeutic efficacy and antimetastatic potential. This study showcases the potential of short-peptide-based nanocarriers for the design and development of stable and efficient photothermal platforms. The multifaceted therapeutic strategy, which merges photothermal ablation with vascular disruption and hypoxia-activated chemotherapy, holds great promise for advancing the efficacy and scope of cancer treatment modalities.


Subject(s)
Melanoma , Animals , Mice , Melanoma/pathology , Melanoma/drug therapy , Melanoma/metabolism , Cell Line, Tumor , Humans , Photothermal Therapy , Nanoparticles/chemistry , Peptides/chemistry , Peptides/pharmacology , Stilbenes/chemistry , Stilbenes/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Tumor Hypoxia/drug effects , Tirapazamine/chemistry , Tirapazamine/pharmacology , Indocyanine Green/analogs & derivatives
16.
J Med Chem ; 67(15): 13491-13506, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39069676

ABSTRACT

Prostate-specific membrane antigen (PSMA) is an excellent target for cancer detection and therapy. Hypoxia is prevalent in solid tumors, and various nitroimidazole (NI) radioligands can be trapped inside hypoxic cells for diagnosis and therapy. To enhance tumor uptake and retention, we designed bivalent agents (compounds 1-8) incorporating a hypoxia-sensitive NI-moiety and a PSMA-targeting group. Ligands 1-8 were successfully prepared and labeled with 68Ga or 177Lu. Among them, [68Ga]Ga-8 ([68Ga]Ga-AAZTA-NI-PSMA-093) demonstrated significantly higher cellular accumulation under hypoxic conditions than under normoxic conditions, suggesting hypoxia-selective trapping by the introduction of NI group. PET/CT imaging at 60 min postinjection of [68Ga]Ga-8 revealed high tumor uptake (SUVmax: 10.68%ID/mL) in the tumor-bearing mice model. SPECT/CT imaging of [177Lu]Lu-8 at 24 and 48 h postinjection demonstrated excellent accumulation and retention. Preliminary studies indicate that [68Ga]Ga/[177Lu]Lu-8 may be promising bivalent agents targeting hypoxia and PSMA binding for diagnosis and radiotherapy.


Subject(s)
Antigens, Surface , Gallium Radioisotopes , Glutamate Carboxypeptidase II , Lutetium , Prostatic Neoplasms , Male , Gallium Radioisotopes/chemistry , Animals , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Humans , Glutamate Carboxypeptidase II/metabolism , Mice , Antigens, Surface/metabolism , Lutetium/chemistry , Radioisotopes/chemistry , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Cell Line, Tumor , Tissue Distribution , Mice, Nude , Tumor Hypoxia
17.
J Colloid Interface Sci ; 676: 506-520, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39047378

ABSTRACT

The synergistic anti-tumor impact of phototherapy and a cascading immune response are profoundly limited by hypoxia and a weakened immune response. Intravenous and intratumoral injection of therapeutic drugs also cause pain, rapid drug clearance and low utilization rates. Here, a novel cryo-millineedle platform for intratumoral delivery of a phototherapy system, S.epi@IR820, is developed in this work, combining the properties of Staphylococcus epidermidis (S. epidermidis) and IR820 for photo-immunotherapy of colorectal cancer. In this cryo-millineedle platform, S. epidermidis enhances the near-infrared absorption and light stability of IR820 and catalyzes the decomposition of H2O2 into O2 via an endogenous catalase to relieve tumor hypoxia, improve phototherapy and enhance immunogenic cell death (ICD). More interestingly, the native immunogenicity of S. epidermidis and ICD elicited by phototherapy achieved a potent anti-tumor immune response. To the best of our knowledge, this is the first study to utilize native S. epidermidis to relieve hypoxia and facilitate phototherapy. Both in vitro and in vivo experiments showed that the millineedle based phototherapy system can efficiently catalyse the decomposition of H2O2 into O2, facilitate phototherapeutic killing of CT26 tumor cells by S.epi@IR820 and enhance ICD, thus successfully activated the immune response and achieved the photo-immunotherapy against colorectal cancer. In conclusion, this study provides a novel strategy for enhanced anti-tumor efficiency of photo-immunotherapy, and develops an effective method for orthotopic administration of tumors.


Subject(s)
Catalase , Colorectal Neoplasms , Immunotherapy , Staphylococcus epidermidis , Colorectal Neoplasms/therapy , Colorectal Neoplasms/pathology , Catalase/chemistry , Catalase/metabolism , Mice , Animals , Phototherapy , Humans , Tumor Hypoxia/drug effects , Cell Line, Tumor , Particle Size , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Mice, Inbred BALB C , Cell Proliferation/drug effects , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Cell Survival/drug effects
18.
Colloids Surf B Biointerfaces ; 241: 114070, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38968858

ABSTRACT

Reactive oxygen species (ROS)-mediated therapeutic strategies, including chemodynamic therapy (CDT), photodynamic therapy (PDT), and their combination, are effective for treating cancer. Developing a nanoreactor with combined functions of catalase (CAT) and peroxidase (POD) that can simultaneously convert excess H2O2 in tumors into O2 required for type II PDT and hydroxyl radicals (•OH) for CDT can help achieve combined therapy. Here, we reported on a safe Fe2O3/CNx nanoreactor with dual enzyme simulated activity, in which CNx sheet was the carrier and reducing agent to convert Fe2O3 to Fe2+. After modified by MgO2 and photosensitizer Ce6, MgO2-Fe2O3/CNx-Ce6 (MFCC) platform integrated multiple functions, including photosensitizer delivery, compensated H2O2 continuous supply, relieve of hypoxia, generation of •OH and consumption of GSH into a single formulation. Under 660 nm irradiation for 4 min, MFCC actives more ROS to conduct PDT/CDT, leading to the remarkable reduced survival rate of breast cancer cells to 14 %. Due to the enhanced permeability and retention (EPR) effect, MFCC can retain and accumulate at the tumor site of mice for a longer period that inhibit the expression of tumor angiogenic factors, suppress tumor neovascularization, and suppress the proliferation and growth of tumor cells.


Subject(s)
Ferric Compounds , Photochemotherapy , Photosensitizing Agents , Tumor Hypoxia , Animals , Humans , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Tumor Hypoxia/drug effects , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Female , Reactive Oxygen Species/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Mice, Inbred BALB C , Cell Line, Tumor , Hydrogen Peroxide/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Particle Size
19.
Front Immunol ; 15: 1384249, 2024.
Article in English | MEDLINE | ID: mdl-38994360

ABSTRACT

Glioblastoma (GBM) tumors are the most aggressive primary brain tumors in adults that, despite maximum treatment, carry a dismal prognosis. GBM tumors exhibit tissue hypoxia, which promotes tumor aggressiveness and maintenance of glioma stem cells and creates an overall immunosuppressive landscape. This article reviews how hypoxic conditions overlap with inflammatory responses, favoring the proliferation of immunosuppressive cells and inhibiting cytotoxic T cell development. Immunotherapies, including vaccines, immune checkpoint inhibitors, and CAR-T cell therapy, represent promising avenues for GBM treatment. However, challenges such as tumor heterogeneity, immunosuppressive TME, and BBB restrictiveness hinder their effectiveness. Strategies to address these challenges, including combination therapies and targeting hypoxia, are actively being explored to improve outcomes for GBM patients. Targeting hypoxia in combination with immunotherapy represents a potential strategy to enhance treatment efficacy.


Subject(s)
Brain Neoplasms , Glioblastoma , Tumor Microenvironment , Humans , Glioblastoma/immunology , Glioblastoma/therapy , Glioblastoma/pathology , Tumor Microenvironment/immunology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Animals , Immunotherapy/methods , Tumor Hypoxia
20.
Cell Death Dis ; 15(7): 503, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003252

ABSTRACT

Glioblastoma multiforme (GBM) is the most common adult primary brain tumor. The standard clinical treatment of GBM includes a maximal surgical resection followed by concomitant radiotherapy (RT) and chemotherapy sessions with Temozolomide (TMZ) in addition to adjuvant TMZ cycles. Despite the severity of this protocol, GBM is highly resistant and recurs in almost all cases while the protocol remains unchanged since 2005. Limited-diffusion or chronic hypoxia has been identified as one of the major key players driving this aggressive phenotype. The presence of hypoxia within the tumor bulk contributes to the activation of hypoxia signaling pathway mediated by the hypoxia-inducing factors (HIFs), which in turn activate biological mechanisms to ensure the adaptation and survival of GBM under limited oxygen and nutrient supply. Activated downstream pathways are involved in maintaining stem cell-like phenotype, inducing mesenchymal shift, invasion, and migration, altering the cellular and oxygen metabolism, and increasing angiogenesis, autophagy, and immunosuppression. Therefore, in this review will discuss the recent preclinical and clinical approaches that aim at targeting tumor hypoxia to enhance the response of GBM to conventional therapies along with their results and limitations upon clinical translation.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/metabolism , Glioblastoma/pathology , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Animals , Cell Hypoxia , Tumor Hypoxia , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL