Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 360
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Int J Nanomedicine ; 19: 6499-6513, 2024.
Article in English | MEDLINE | ID: mdl-38946887

ABSTRACT

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


Subject(s)
Nanoparticles , Photothermal Therapy , Polylactic Acid-Polyglycolic Acid Copolymer , Reactive Oxygen Species , Animals , Photothermal Therapy/methods , Reactive Oxygen Species/metabolism , Nanoparticles/chemistry , Cell Line, Tumor , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Mice , Indoles/pharmacology , Indoles/chemistry , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/radiotherapy , Neoplasms/therapy , Neoplasms/metabolism , Nanomedicine
13.
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
14.
Dalton Trans ; 53(27): 11242-11246, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38919991

ABSTRACT

Herein, we report a composite COF material loaded with a Pt nanoenzyme and an organic photosensitizer BODIPY, synthesized via a stepwise post-synthetic modification. The obtained Pt@COF-BDP nanoparticles can efficiently and continuously convert H2O2 to O2, thereby increasing the efficiency of single-linear oxygen production and achieving efficient tumor inhibition.


Subject(s)
Boron Compounds , Metal-Organic Frameworks , Photochemotherapy , Photosensitizing Agents , Platinum , Boron Compounds/chemistry , Boron Compounds/pharmacology , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Platinum/chemistry , Platinum/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Metal-Organic Frameworks/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Mice , Nanoparticles/chemistry , Tumor Hypoxia/drug effects , Hydrogen Peroxide/chemistry
15.
Chin J Integr Med ; 30(9): 799-808, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38850481

ABSTRACT

OBJECTIVE: To investigate whether Buthus martensii karsch (Scorpiones), Scolopendra subspinipes mutilans L. Koch (Scolopendra) and Gekko gecko Linnaeus (Gekko) could ameliorate the hypoxic tumor microenvironment and inhibit lung cancer growth and metastasis by regulating phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin/hypoxia-inducible factor-1α (PI3K/AKT/mTOR/HIF-1α) signaling pathway. METHODS: Male C57BL/6J mice were inoculated with luciferase labeled LL/2-luc-M38 cell suspension to develop lung cancer models, with rapamycin and cyclophosphamide as positive controls. Carboxy methyl cellulose solutions of Scorpiones, Scolopendra and Gekko were administered intragastrically as 0.33, 0.33, and 0.83 g/kg, respectively once daily for 21 days. Fluorescent expression were detected every 7 days after inoculation, and tumor growth curves were plotted. Immunohistochemistry was performed to determine CD31 and HIF-1α expressions in tumor tissue and microvessel density (MVD) was analyzed. Western blot was performed to detect the expression of PI3K/AKT/mTOR/HIF-1α signaling pathway-related proteins. Enzyme-linked immunosorbent assay was performed to detect serum basic fibroblast growth factor (bFGF), transforming growth factor-ß1 (TGF-ß1) and vascular endothelial growth factor (VEGF) in mice. RESULTS: Scorpiones, Scolopendra and Gekko prolonged the survival time and inhibited lung cancer metastasis and expression of HIF-1α (all P<0.01). Moreover, Scorpiones, Scolopendra and Gekko inhibited the phosphorylation of AKT and ribosomal protein S6 kinase (p70S6K) (P<0.05 or P<0.01). In addition, they also decreased the expression of CD31, MVD, bFGF, TGF-ß1 and VEGF compared with the model group (P<0.05 or P<0.01). CONCLUSION: Scorpiones, Scolopendra and Gekko all showed beneficial effects on lung cancer by ameliorating the hypoxic tumor microenvironment via PI3K/AKT/mTOR/HIF-1α signaling pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Lung Neoplasms , Mice, Inbred C57BL , Neoplasm Metastasis , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Tumor Microenvironment , Animals , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Male , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Mice , Neovascularization, Pathologic , Tumor Hypoxia/drug effects
16.
Sci Rep ; 14(1): 8532, 2024 06 03.
Article in English | MEDLINE | ID: mdl-38830912

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) poses challenges due to late-stage diagnosis and limited treatment response, often attributed to the hypoxic tumor microenvironment (TME). Sonoporation, combining ultrasound and microbubbles, holds promise for enhancing therapy. However, additional preclinical research utilizing commercially available ultrasound equipment for PDAC treatment while delving into the TME's intricacies is necessary. This study investigated the potential of using a clinically available ultrasound system and phase 2-proven microbubbles to relieve tumor hypoxia and enhance the efficacy of chemotherapy and immunotherapy in a murine PDAC model. This approach enables early PDAC detection and blood-flow-sensitive Power-Doppler sonoporation in combination with chemotherapy. It significantly extended treated mice's median survival compared to chemotherapy alone. Mechanistically, this combination therapy enhanced tumor perfusion and substantially reduced tumor hypoxia (77% and 67%, 1- and 3-days post-treatment). Additionally, cluster of differentiation 8 (CD8) T-cell infiltration increased four-fold afterward. The combined treatment demonstrated a strengthening of the anti-programmed death-ligand 1(αPDL1) therapy against PDAC. Our study illustrates the feasibility of using a clinically available ultrasound system with NH-002 microbubbles for early tumor detection, alleviating hypoxic TME, and improving chemotherapy and immunotherapy. It suggests the development of an adjuvant theragnostic protocol incorporating Power-Doppler sonoporation for pancreatic tumor treatment.


Subject(s)
Carcinoma, Pancreatic Ductal , Immunotherapy , Microbubbles , Pancreatic Neoplasms , Tumor Microenvironment , Animals , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Mice , Immunotherapy/methods , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Tumor Microenvironment/drug effects , Cell Line, Tumor , Tumor Hypoxia/drug effects , Combined Modality Therapy , Humans , Female
17.
Biomaterials ; 310: 122634, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38823195

ABSTRACT

The hypoxic nature of pancreatic cancer, one of the most lethal malignancies worldwide, significantly impedes the effectiveness of chemoradiotherapy. Although the development of oxygen carriers and hypoxic sensitizers has shown promise in overcoming tumor hypoxia. The heterogeneity of hypoxia-primarily caused by limited oxygen penetration-has posed challenges. In this study, we designed a hypoxia-responsive nano-sensitizer by co-loading tirapazamine (TPZ), KP372-1, and MK-2206 in a metronidazole-modified polymeric vesicle. This nano-sensitizer relies on efficient endogenous NAD(P)H quinone oxidoreductase 1-mediated redox cycling induced by KP372-1, continuously consuming periphery oxygen and achieving evenly distributed hypoxia. Consequently, the normalized tumor microenvironment facilitates the self-amplified release and activation of TPZ without requiring deep penetration. The activated TPZ and metronidazole further sensitize radiotherapy, significantly reducing the radiation dose needed for extensive cell damage. Additionally, the coloaded MK-2206 complements inhibition of therapeutic resistance caused by Akt activation, synergistically enhancing the hypoxic chemoradiotherapy. This successful hypoxia normalization strategy not only overcomes hypoxia resistance in pancreatic cancer but also provides a potential universal approach to sensitize hypoxic tumor chemoradiotherapy by reshaping the hypoxic distribution.


Subject(s)
Chemoradiotherapy , Drug Liberation , Pancreatic Neoplasms , Tirapazamine , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Humans , Tirapazamine/pharmacology , Chemoradiotherapy/methods , Cell Line, Tumor , Animals , Mice, Nude , Heterocyclic Compounds, 3-Ring/pharmacology , Nanoparticles/chemistry , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Tumor Hypoxia/drug effects , Mice, Inbred BALB C , Metronidazole/pharmacology , Metronidazole/therapeutic use , Tumor Microenvironment/drug effects
18.
J Nanobiotechnology ; 22(1): 374, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926723

ABSTRACT

BACKGROUND: Hypoxia-activated prodrugs present new opportunities for safe and effective tumor drug resistance therapy due to their high selectivity for hypoxic cells. However, the uneven distribution of oxygen in solid tumor and insufficient hypoxia in the tumor microenvironment greatly limit its therapeutic efficacy. RESULTS: In this paper, a novel AQ4N-Mn(II)@PDA coordination nanoplatform was designed and functionalized with GMBP1 to target drug-resistant tumor cells. Its excellent photothermal conversion efficiency could achieve local high-temperature photothermal therapy in tumors, which could not only effectively exacerbate tumor hypoxia and thus improve the efficacy of hypoxia-activated chemotherapy of AQ4N but also significantly accelerate Mn2+-mediated Fenton-like activity to enhance chemodynamic therapy. Moreover, real-time monitoring of blood oxygen saturation through photoacoustic imaging could reflect the hypoxic status of tumors during treatment. Furthermore, synergistic treatment effectively inhibited tumor growth and improved the survival rate of mice bearing orthotopic drug-resistant tumors. CONCLUSIONS: This study not only provided a new idea for PTT combined with hypoxia-activated chemotherapy and CDT for drug-resistant tumors but also explored a vital theory for real-time monitoring of hypoxia during treatment.


Subject(s)
Drug Resistance, Neoplasm , Photothermal Therapy , Animals , Mice , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Humans , Photothermal Therapy/methods , Mice, Inbred BALB C , Nanoparticles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Tumor Microenvironment/drug effects , Mice, Nude , Prodrugs/pharmacology , Prodrugs/chemistry , Tumor Hypoxia/drug effects , Manganese/chemistry , Female , Neoplasms/drug therapy , Anthraquinones
19.
BMC Vet Res ; 20(1): 196, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741109

ABSTRACT

BACKGROUND: Hypoxia is a detrimental factor in solid tumors, leading to aggressiveness and therapy resistance. OMX, a tunable oxygen carrier from the heme nitric oxide/oxygen-binding (H-NOX) protein family, has the potential to reduce tumor hypoxia. [18F]Fluoromisonidazole ([18F]FMISO) positron emission tomography (PET) is the most widely used and investigated method for non-invasive imaging of tumor hypoxia. In this study, we used [18F]FMISO PET/CT (computed tomography) to assess the effect of OMX on tumor hypoxia in spontaneous canine tumors. RESULTS: Thirteen canine patients with various tumors (n = 14) were randomly divided into blocks of two, with the treatment groups alternating between receiving intratumoral (IT) OMX injection (OMX IT group) and intravenous (IV) OMX injection (OMX IV group). Tumors were regarded as hypoxic if maximum tumor-to-muscle ratio (TMRmax) was greater than 1.4. In addition, hypoxic volume (HV) was defined as the region with tumor-to-muscle ratio greater than 1.4 on [18F]FMISO PET images. Hypoxia was detected in 6/7 tumors in the OMX IT group and 5/7 tumors in the OMX IV injection group. Although there was no significant difference in baseline hypoxia between the OMX IT and IV groups, the two groups showed different responses to OMX. In the OMX IV group, hypoxic tumors (n = 5) exhibited significant reductions in tumor hypoxia, as indicated by decreased TMRmax and HV in [18F]FMISO PET imaging after treatment. In contrast, hypoxic tumors in the OMX IT group (n = 6) displayed a significant increase in [18F]FMISO uptake and variable changes in TMRmax and HV. CONCLUSIONS: [18F]FMISO PET/CT imaging presents a promising non-invasive procedure for monitoring tumor hypoxia and assessing the efficacy of hypoxia-modulating therapies in canine patients. OMX has shown promising outcomes in reducing tumor hypoxia, especially when administered intravenously, as evident from reductions in both TMRmax and HV in [18F]FMISO PET imaging.


Subject(s)
Dog Diseases , Misonidazole , Neoplasms , Positron Emission Tomography Computed Tomography , Tumor Hypoxia , Animals , Dogs , Misonidazole/analogs & derivatives , Positron Emission Tomography Computed Tomography/veterinary , Positron Emission Tomography Computed Tomography/methods , Dog Diseases/diagnostic imaging , Dog Diseases/drug therapy , Female , Tumor Hypoxia/drug effects , Male , Neoplasms/veterinary , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Thiosemicarbazones/therapeutic use , Thiosemicarbazones/pharmacology , Coordination Complexes
20.
ACS Biomater Sci Eng ; 10(6): 4009-4017, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38722972

ABSTRACT

It still remains challenging to design multifunctional therapeutic reagents for effective cancer therapy under a unique tumor microenvironment including insufficient endogenous H2O2 and O2, low pH, and a high concentration of glutathione (GSH). In this work, a CO-based phototherapeutic system triggered by photogenerated holes, which consisted of ionic liquid (IL), the CO prodrug Mn2(CO)10, and iridium(III) porphyrin (IrPor) modified carbonized ZIF-8-doped graphitic carbon nitride nanocomposite (IL/ZCN@Ir(CO)), was designed for cascade hypoxic tumors. Upon light irradiation, the photogenerated holes on IL/ZCN@Ir(CO) oxidize water into H2O2, which subsequently induces Mn2(CO)10 to release CO. Meanwhile, IrPor can convert H2O2 to hydroxyl radical (•OH) and subsequent singlet oxygen (1O2), which further triggers CO release. Moreover, the degraded MnO2 shows activity for glutathione (GSH) depletion and mimics peroxidase, leading to GSH reduction and •OH production in tumors. Thus, this strategy can in situ release high concentrations of CO and reactive oxygen species (ROS) and deplete GSH to efficiently induce cell apoptosis under hypoxic conditions, which has a high inhibiting effect on the growth of tumors, offering an attractive strategy to amplify CO and ROS generation to meet therapeutic requirements in cancer treatment.


Subject(s)
Carbon Monoxide , Glutathione , Carbon Monoxide/metabolism , Carbon Monoxide/chemistry , Carbon Monoxide/pharmacology , Humans , Glutathione/metabolism , Glutathione/chemistry , Animals , Cell Line, Tumor , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/radiotherapy , Tumor Hypoxia/drug effects , Mice , Iridium/chemistry , Iridium/pharmacology , Graphite/chemistry , Graphite/pharmacology , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Nitrogen Compounds
SELECTION OF CITATIONS
SEARCH DETAIL