Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 337
Filter
1.
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
2.
Acta Biomater ; 181: 402-414, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38734282

ABSTRACT

Tumor hypoxia, high oxidative stress, and low immunogenic create a deep-rooted immunosuppressive microenvironment, posing a major challenge to the therapeutic efficiency of cancer immunotherapy for solid tumor. Herein, an intelligent nanoplatform responsive to the tumor microenvironment (TME) capable of hypoxia relief and immune stimulation has been engineered for efficient solid tumor immunotherapy. The MnO2@OxA@OMV nanoreactor, enclosing bacterial-derived outer membrane vesicles (OMVs)-wrapped MnO2 nanoenzyme and the immunogenic cell death inducer oxaliplatin (OxA), demonstrated intrinsic catalase-like activity within the TME, which effectively catalyzed the endogenous H2O2 into O2 to enable a prolonged oxygen supply, thereby alleviating the tumor's oxidative stress and hypoxic TME, and expediting OxA release. The combinational action of OxA-caused ICD effect and Mn2+ from nanoreactor enabled the motivation of the cGAS-STING pathway to significantly improve the activation of STING and dendritic cells (DCs) maturation, resulting in metalloimmunotherapy. Furthermore, the immunostimulant OMVs played a crucial role in promoting the infiltration of activated CD8+T cells into the solid tumor. Overall, the nanoreactor offers a robust platform for solid tumor treatment, highlighting the significant potential of combining relief from tumor hypoxia and immune stimulation for metalloimmunotherapy. STATEMENT OF SIGNIFICANCE: A tailor-made nanoreactor was fabricated by enclosing bacterial-derived outer membrane vesicles (OMVs) onto MnO2 nanoenzyme and loading with immunogenic cell death inducer oxaliplatin (OxA) for tumor metalloimmunotherapy. The nanoreactor possesses intrinsic catalase-like activity within the tumor microenvironment, which effectively enabled a prolonged oxygen supply by catalyzing the conversion of endogenous H2O2 into O2, thereby alleviating tumor hypoxia and expediting OxA release. Furthermore, the TME-responsive release of nutritional Mn2+ sensitized the cGAS-STING pathway and collaborated with OxA-induced immunogenic cell death (ICD). Combing with immunostimulatory OMVs enhances the uptake of nanoreactors by DCs and promotes the infiltration of activated CD8+T cells. This nanoreactor offers a robust platform for solid tumor treatment, highlighting the significant potential of combining relief from tumor hypoxia and immune stimulation for metalloimmunotherapy.


Subject(s)
Immunotherapy , Tumor Microenvironment , Animals , Immunotherapy/methods , Mice , Tumor Microenvironment/drug effects , Cell Line, Tumor , Tumor Hypoxia/drug effects , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Oxaliplatin/pharmacology , Oxaliplatin/chemistry , Oxides/chemistry , Oxides/pharmacology , Manganese/chemistry , Manganese/pharmacology , Humans , Female , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/immunology , Neoplasms/drug therapy , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/immunology , Mice, Inbred C57BL
3.
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
4.
J Colloid Interface Sci ; 666: 244-258, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38598997

ABSTRACT

Starvation therapy has shown promise as a cancer treatment, but its efficacy is often limited when used alone. In this work, a multifunctional nanoscale cascade enzyme system, named CaCO3@MnO2-NH2@GOx@PVP (CMGP), was fabricated for enhanced starvation/chemodynamic combination cancer therapy. CMGP is composed of CaCO3 nanoparticles wrapped in a MnO2 shell, with glucose oxidase (GOx) adsorbed and modified with polyvinylpyrrolidone (PVP). MnO2 decomposes H2O2 in cancer cells into O2, which enhances the efficiency of GOx-mediated starvation therapy. CaCO3 can be decomposed in the acidic cancer cell environment, causing Ca2+ overload in cancer cells and inhibiting mitochondrial metabolism. This synergizes with GOx to achieve more efficient starvation therapy. Additionally, the H2O2 and gluconic acid produced during glucose consumption by GOx are utilized by MnO2 with catalase-like activity to enhance O2 production and Mn2+ release. This process accelerates glucose consumption, reactive oxygen species (ROS) generation, and CaCO3 decomposition, promoting the Ca2+ release. CMGP can alleviate tumor hypoxia by cycling the enzymatic cascade reaction, which increases enzyme activity and combines with Ca2+ overload to achieve enhanced combined starvation/chemodynamic therapy. In vitro and in vivo studies demonstrate that CMGP has effective anticancer abilities and good biosafety. It represents a new strategy with great potential for combined cancer therapy.


Subject(s)
Calcium Carbonate , Glucose Oxidase , Manganese Compounds , Oxides , Glucose Oxidase/metabolism , Glucose Oxidase/chemistry , Glucose Oxidase/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Oxides/chemistry , Oxides/pharmacology , Humans , Animals , Calcium Carbonate/chemistry , Calcium Carbonate/pharmacology , Calcium Carbonate/metabolism , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Povidone/chemistry , Povidone/pharmacology , Tumor Hypoxia/drug effects , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Particle Size , Cell Line, Tumor , Hydrogen Peroxide/metabolism , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Surface Properties , Mice, Inbred BALB C
5.
Bioorg Med Chem Lett ; 106: 129773, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38677561

ABSTRACT

Hypoxia is a common phenomenon in solid tumors, and its presence inhibits the efficacy of tumor chemotherapy and radiotherapy. Accurate measurement of hypoxia before tumor treatment is essential. Three propylene amine oxime (PnAO) derivatives with different substituents attached to 2-nitroimidazole were synthesized in the work, they are 3,3,9,9-tetramethyl-1,11-bis(4-bromo-2-nitro-1H-imidazol-1-yl)-4,8-diazaundecane-2,10-dione dioxime (Br2P2), 3,3,9,9-tetramethyl-1,11-bis(4-methyl-2-nitro-1H-imidazol-1-yl)-4,8-diazaundecane-2,10-dione dioxime (Me2P2) and 3,3,9,9-tetramethyl-1,11-bis(4,5-dimethyl-2-nitro-1H-imidazol-1-yl)-4,8-diazaundecane-2,10-dione dioxime (2Me2P2). The three compounds were radiolabeled with 99mTc to give three complexes([99mTc]Tc-Br2P2, [99mTc]Tc-Me2P2 and [99mTc]Tc-2Me2P2) with good in vitro stability. [99mTc]Tc-Me2P2 with a more suitable reduction potential had the highest hypoxic cellular uptake, compared with [99mTc]Tc-2P2 that have been previously reported, [99mTc]Tc-Br2P2 and [99mTc]Tc-2Me2P2. Biodistribution results in S180 tumor-bearing mice demonstrated that [99mTc]Tc-Me2P2 had the highest tumor-to-muscle (T/M) ratio (12.37 ± 1.16) at 2 h in the four complexes. Autoradiography and immunohistochemical staining results revealed that [99mTc]Tc-Me2P2 specifically targeted tumor hypoxic regions. The SPECT/CT imaging results showed that [99mTc]Tc-Me2P2 could target the tumor site. [99mTc]Tc-Me2P2 may become a potential hypoxia imaging agent.


Subject(s)
Nitroimidazoles , Organotechnetium Compounds , Oximes , Tumor Hypoxia , Oximes/chemistry , Oximes/chemical synthesis , Nitroimidazoles/chemistry , Nitroimidazoles/chemical synthesis , Animals , Mice , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/chemical synthesis , Tumor Hypoxia/drug effects , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacology , Humans , Tissue Distribution , Molecular Structure , Cell Line, Tumor , Structure-Activity Relationship
6.
Asian Pac J Cancer Prev ; 25(4): 1315-1324, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38679992

ABSTRACT

OBJECTIVE: Tumor hypoxia induces the production of Hypoxia-Inducible Factor (HIF)-1 alpha, which interacts with NF-kB, leading to cancer proliferation and metastasis. This study investigated the effect of tumor hypoxia modulation using carbogen (95% O2 and 5% CO2) and nicotinamide on reducing soluble interleukin-2 receptor (sIL-2R) levels in newly diagnosed DLBCL patients with tissue overexpression of HIF-1α ≥10%. MATERIAL AND METHODS: A prospective randomized controlled clinical trial was conducted at Dr. Kariadi Hospital in Semarang, Indonesia, from 2021 to 2022. Newly diagnosed DLBCL patients with tissue HIF-1α ≥10% were randomized into an intervention group (nicotinamide 2,000 mg + carbogen 10 liters/min during R-CHOP) and a control group (R-CHOP alone) for one cycle. sIL-2R levels were measured in the blood before and after intervention. RESULTS: The intervention group showed a significant reduction in sIL-2R levels after chemotherapy (p=0.026), with 85% of samples exhibiting a decrease. In contrast, only 45% of samples in the control group demonstrated a decrease in sIL-2R levels (p=0.184). The median sIL-2R level decreased from 139.50 pg/mL to 70.50 pg/mL in the intervention group, while the control group exhibited an increase from 182.50 pg/mL to 250.00 pg/mL following one cycle of chemotherapy. CONCLUSION: Tumor hypoxia modulation led to a significant decrease in serum sIL-2R levels, potentially through improvements in the crosstalk between hypoxia and inflammation pathways.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Cyclophosphamide , Doxorubicin , Lymphoma, Large B-Cell, Diffuse , Receptors, Interleukin-2 , Tumor Hypoxia , Vincristine , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/metabolism , Male , Female , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Middle Aged , Tumor Hypoxia/drug effects , Prospective Studies , Receptors, Interleukin-2/blood , Receptors, Interleukin-2/metabolism , Vincristine/therapeutic use , Doxorubicin/therapeutic use , Cyclophosphamide/therapeutic use , Adult , Prednisone/therapeutic use , Prognosis , Rituximab/therapeutic use , Follow-Up Studies , Aged , Indonesia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/blood
7.
Chem Commun (Camb) ; 60(40): 5322-5325, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38666540

ABSTRACT

A small molecule-based NIR-II type-I photosensitizer (IT-IC) with a strong push-pull effect and good planar π-conjugated structure was synthesized. The IT-IC NPs exhibited strong light absorption, outstanding NIR-II fluorescence emission, excellent photothermal conversion and efficient type-I/II ROS generation, showing encouraging therapeutic outcomes for hypoxic tumors.


Subject(s)
Infrared Rays , Photosensitizing Agents , Theranostic Nanomedicine , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Humans , Animals , Mice , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Reactive Oxygen Species/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Molecular Structure , Photochemotherapy , Tumor Hypoxia/drug effects , Cell Survival/drug effects , Nanoparticles/chemistry
8.
ACS Nano ; 18(19): 12261-12275, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38683132

ABSTRACT

Cancer immunotherapy holds significant promise for addressing diverse malignancies. Nevertheless, its efficacy remains constrained by the intricate tumor immunosuppressive microenvironment. Herein, a light-triggered nanozyme Fe-TCPP-R848-PEG (Fe-MOF-RP) was designed for remodeling the immunosuppressive microenvironment. The Fe-TCPP-MOFs were utilized not only as a core catalysis component against tumor destruction but also as a biocompatible delivery vector of an immunologic agonist, improving its long circulation and tumor enrichment. Concurrently, it catalyzes the decomposition of H2O2 within the tumor, yielding oxygen to augment photodynamic therapy. The induced ferroptosis, in synergy with photodynamic therapy, prompts the liberation of tumor-associated antigens from tumor cells inducing immunogenic cell death. Phototriggered on-demand release of R848 agonists stimulated the maturation of dendritic cells and reverted the tumor-promoting M2 phenotypes into adoptive M1 macrophages, which further reshaped the tumor immunosuppressive microenvironment. Notably, the nanozyme effectively restrains well-established tumors, such as B16F10 melanoma. Moreover, it demonstrates a distal tumor-inhibiting effect upon in situ light treatment. What is more, in a lung metastasis model, it elicits robust immune memory, conferring enduring protection against tumor rechallenge. Our study presents a straightforward and broadly applicable strategy for crafting nanozymes with the potential to effectively thwart cancer recurrence and metastasis.


Subject(s)
Ferroptosis , Light , Tumor Microenvironment , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Ferroptosis/drug effects , Mice , Mice, Inbred C57BL , Photochemotherapy , Tumor Hypoxia/drug effects , Nanoparticles/chemistry , Immunotherapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Cell Line, Tumor
9.
Biomater Sci ; 12(11): 2831-2840, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38683541

ABSTRACT

Photodynamic therapy (PDT) is a minimally invasive therapeutic modality employed for the treatment of various types of cancers, localized infections, and other diseases. Upon illumination, the photo-excited photosensitizer generates singlet oxygen and other reactive species, thereby inducing cytotoxicity in the target cells. The hypoxic tumour microenvironment (TME), however, poses a limitation on the supply of oxygen in tumour tissues. Moreover, under such conditions, tumour metastasis and drug resistance frequently occur, further compromising the efficacy of PDT in combating tumours. Traditionally, type I photosensitizers with lower oxygen consumption demonstrate significant potential in overcoming hypoxic environments and play a crucial role in determining the therapeutic efficacy of PDT because type I photosensitizers can generate highly cytotoxic free radicals. In comparison, type II photosensitizers exhibit high oxygen dependence. The rate of reactive oxygen species (ROS) generation in the type II process is significantly higher than that in the type I process. Thus, the efficiency and selectivity of PDT depend on the properties of the photosensitizer. Here, the recent development and application of type I and type II photosensitizers, mainly in the past year, are summarized. The design methods, electronic structures, photophysical properties, lipophilic properties, electric charge, and other molecular characteristics of these photosensitizers are discussed in detail. These modifications alter the microstructure of photosensitizers and directly impact the results of PDT. The main content of this paper will have a positive promoting and inspiring effect on the future development of PDT.


Subject(s)
Neoplasms , Photochemotherapy , Photosensitizing Agents , Reactive Oxygen Species , Tumor Microenvironment , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Tumor Microenvironment/drug effects , Humans , Reactive Oxygen Species/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Tumor Hypoxia/drug effects
10.
J Am Chem Soc ; 146(20): 13805-13816, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38552185

ABSTRACT

Cuproptosis, a copper-dependent cell death process, has been confirmed to further activate the immune response and mediate the immune resistance. However, hypoxic tumor microenvironment hampers cuproptosis sensitivity and suppresses the body's antitumor immune response. Herein, we have successfully immobilized and functionalized catalase (CAT) with long single-stranded DNA containing polyvalent CpG sequences through rolling circle amplification (RCA) techniques, obtaining an enzyme-cored spherical nucleic acid nanoplatform (CAT-ecSNA-Cu) to deliver copper ions for cuproptosis. The presence of long-stranded DNA-protected CAT enhances mitochondrial respiration by catalyzing the conversion of H2O2 to O2, thereby sensitizing cuproptosis. Meanwhile, increased tumor oxygenation suppresses the expression of the hypoxia-inducible factor-1 (HIF-1) protein, resulting in the alleviation of the immunosuppressive tumor microenvironment. Of note, cuproptosis induces immunogenic cell death (ICD), which facilitates dendritic cell (DC) maturation and enhances antigen presentation through polyCpG-supported Toll-like receptor 9 (TLR9) activation. Furthermore, cuproptosis-induced PD-L1 upregulation in tumor cells complements checkpoint blockers (αPD-L1), enhancing antitumor immunity. The strategy of enhancing cuproptosis-mediated antitumor immune responses by alleviating hypoxia effectively promotes the activation and proliferation of effector T cells, ultimately leading to long-term immunity against cancer.


Subject(s)
Catalase , Copper , Tumor Hypoxia , Tumor Hypoxia/drug effects , Animals , Copper/chemistry , Catalase/metabolism , Catalase/chemistry , Mice , Tumor Microenvironment/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Immunogenic Cell Death/drug effects , Dendritic Cells/immunology , Dendritic Cells/drug effects
11.
Adv Mater ; 36(23): e2310875, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38450765

ABSTRACT

Photodynamic therapy (PDT) has been approved for clinic. However, powerless efficiency for deep hypoxic tumor therapy remains an enormous challenge for PDT. Herein, a hypoxia-sensitive nanotherapeutic system (FTCD-SRGD) based on fullerene (C70) and anoxic activating chemical prodrug tirapazamine (TPZ) is rationally designed for multimodal therapy of deep hypoxic tumors. To enhance the accumulation and achieve specific drug release in tumor, the FTCD-SRGD is modified with cyclo(Arg-Gly-Asp-d-Phe-Lys) (cRGDfK) peptide and disulfide bonds. With the exacerbated hypoxic microenvironment created by C70 consuming O2 for generating reactive oxygen species (ROS), TPZ is activated to produce toxic radical species to ablate deep tumors, which achieves a synergistic treatment of C70-mediated PDT and hypoxia-enhanced chemotherapy. Additionally, given this hypoxia-sensitive system-induced immunogenic cell death (ICD) activating anticancer cytotoxic T lymphocyte to result in more susceptible tumor to immunotherapy, FTCD-SRGD plus immune checkpoint inhibitor (anti-PD-L1) fully inhibit deep hypoxic tumors by promoting infiltration of effector T cells in tumors. Collectively, it is the first time to develop a multimodal therapy system with fullerene-based hypoxia-sensitive PS for deep tumors. The powerful multimodal nanotherapeutic system for combining hypoxia-enhanced PDT and immunotherapy to massacre deep hypoxic tumors can provide a paradigm to combat the present bottleneck of tumor therapy.


Subject(s)
Fullerenes , Photochemotherapy , Photosensitizing Agents , Tirapazamine , Fullerenes/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Animals , Photochemotherapy/methods , Mice , Cell Line, Tumor , Tirapazamine/chemistry , Tirapazamine/pharmacology , Humans , Combined Modality Therapy , Tumor Microenvironment/drug effects , Reactive Oxygen Species/metabolism , Neoplasms/drug therapy , Neoplasms/therapy , Neoplasms/pathology , Tumor Hypoxia/drug effects , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
12.
Photodiagnosis Photodyn Ther ; 46: 104024, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401819

ABSTRACT

This review discusses the possible methods for improving therapeutic efficacies of phthalocyanine (Pcs) -based therapeutic probes in photo- and sono-dynamic therapies under hypoxic conditions. Herein, the structural design strategies including varying the central metal, position substituents and the effects of adjuvant used in supplementing the therapeutics activities of Pcs or formation of NPs are discussed for cancer therapies in hypoxic conditions. Different mechanisms induced for cell death influenced by the compositions of the Pcs-probes are discussed. The focus mainly highlights the oxygen (O2) -dependent mechanisms including methods of supplementing tumour microenvironment O2-concentrations to promote PDT or SDT therapies. Alternatively, O2-independent mechanisms mainly used to evade hypoxia by stimulating anticancer processes that don't require O2 to initiate cell death, such as the Fenton reaction or thermal ablation effects.


Subject(s)
Indoles , Isoindoles , Photochemotherapy , Photosensitizing Agents , Tumor Hypoxia , Indoles/pharmacology , Indoles/therapeutic use , Humans , Isoindoles/pharmacology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photochemotherapy/methods , Tumor Hypoxia/drug effects , Tumor Microenvironment/drug effects
13.
Adv Healthc Mater ; 13(11): e2303856, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38221719

ABSTRACT

Porphyrin photosensitizers are the classic drugs in clinical photodynamic therapy (PDT), but the hypoxia of tumor environment and the rapid oxygen consumption of PDT severely weaken their therapeutic effect. A recently reported water-dependent reversible photoacidity therapy (W-RPAT) is O2-independence, providing a solution for the treatment of hypoxic tumors. In this work, TPP-O-PEG5, a porphyrin derivative with binary properties of PDT and W-RPAT, is designed and synthesized for the first time. The nanoparticles (NPs) of TPP-O-PEG5 encapsulated with DSPE-mPEG2000, an amphiphilic polymer approved by Food and Drug Administration, can simultaneously produce reactive oxygen species and H+ under irradiation of a 660 nm laser, and revert the H+ back under darkness, presenting strong phototoxicity to multiple tumor cell lines with no obvious difference between the IC50 values tested under normoxic (≈20% O2) and hypoxic (<0.5% O2) conditions. Excitingly, in vivo experiments show that the therapeutic effect of TPP-O-PEG5 NPs on large hypoxic tumors is better than that of NPe6, a clinical porphin PDT drug. This work provides a novel strategy for porphyrin photosensitizers to break through the limitation of hypoxic environment, and significantly improve the phototherapeutic effect on hypoxic tumors.


Subject(s)
Photochemotherapy , Photosensitizing Agents , Porphyrins , Photochemotherapy/methods , Porphyrins/chemistry , Porphyrins/pharmacology , Humans , Animals , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Mice , Cell Line, Tumor , Water/chemistry , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , Mice, Inbred BALB C , Tumor Hypoxia/drug effects , Neoplasms/drug therapy , Neoplasms/therapy , Polyethylene Glycols/chemistry , Mice, Nude
14.
Int J Mol Sci ; 23(3)2022 Feb 03.
Article in English | MEDLINE | ID: mdl-35163668

ABSTRACT

Breast cancer is characterized by a hypoxic microenvironment inside the tumor mass, contributing to cell metastatic behavior. Hypoxia induces the expression of hypoxia-inducible factor (HIF-1α), a transcription factor for genes involved in angiogenesis and metastatic behavior, including the vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs), and integrins. Integrin receptors play a key role in cell adhesion and migration, being considered targets for metastasis prevention. We investigated the migratory behavior of hypoxia-cultured triple-negative breast cancer cells (TNBC) and endothelial cells (HUVEC) upon αvß3 integrin blocking with DisBa-01, an RGD disintegrin with high affinity to this integrin. Boyden chamber, HUVEC transmigration, and wound healing assays in the presence of DisBa-01 were performed in hypoxic conditions. DisBa-01 produced similar effects in the two oxygen conditions in the Boyden chamber and transmigration assays. In the wound healing assay, hypoxia abolished DisBa-01's inhibitory effect on cell motility and decreased the MMP-9 activity of conditioned media. These results indicate that αvß3 integrin function in cell motility depends on the assay and oxygen levels, and higher inhibitor concentrations may be necessary to achieve the same inhibitory effect as in normoxia. These versatile responses add more complexity to the role of the αvß3 integrin during tumor progression.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endothelial Cells/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Tumor Hypoxia , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Shape/drug effects , Crotalid Venoms/pharmacology , Culture Media, Conditioned/pharmacology , Disintegrins/pharmacology , Endothelial Cells/pathology , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Matrix Metalloproteinases/metabolism , Neovascularization, Physiologic/drug effects , Oxygen , Protein Subunits/metabolism , Tumor Hypoxia/drug effects
15.
Cells ; 11(2)2022 01 14.
Article in English | MEDLINE | ID: mdl-35053392

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is a highly invasive brain tumour, characterized by its ability to secrete factors promoting its virulence. Brain endothelial cells (BECs) in the GBM environment are physiologically modulated. The present study investigated the modulatory effects of normoxically and hypoxically induced glioblastoma U-87 cell secretions on BECs. METHODS: Conditioned media (CM) were derived by cultivating U-87 cells under hypoxic incubation (5% O2) and normoxic incubation (21% O2). Treated bEnd.3 cells were evaluated for mitochondrial dehydrogenase activity, mitochondrial membrane potential (ΔΨm), ATP production, transendothelial electrical resistance (TEER), and endothelial tight-junction (ETJ) gene expression over 96 h. RESULTS: The coculture of bEnd.3 cells with U-87 cells, or exposure to either hypoxic or normoxic U-87CM, was associated with low cellular viability. The ΔΨm in bEnd.3 cells was hyperpolarized after hypoxic U-87CM treatment (p < 0.0001). However, normoxic U-87CM did not affect the state of ΔΨm. BEC ATP levels were reduced after being cocultured with U-87 cells, or with hypoxic and normoxic CM (p < 0.05). Suppressed mitochondrial activity in bEnd.3 cells was associated with increased transendothelial permeability, while bEnd.3 cells significantly increased the gene expression levels of ETJs (p < 0.05) when treated with U-87CM. CONCLUSIONS: Hypoxic and normoxic glioblastoma paracrine factors differentially suppressed mitochondrial activity in BECs, increasing the BECs' barrier permeability.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Endothelial Cells/pathology , Glioblastoma/pathology , Paracrine Communication , Tumor Hypoxia , Adenosine Triphosphate/metabolism , Animals , Brain Neoplasms/genetics , Cell Line , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Claudin-5/genetics , Claudin-5/metabolism , Coculture Techniques , Culture Media, Conditioned/pharmacology , Electric Impedance , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Occludin/genetics , Occludin/metabolism , Paracrine Communication/drug effects , Paracrine Communication/genetics , Tumor Hypoxia/drug effects , Tumor Hypoxia/genetics
16.
Int J Mol Sci ; 23(2)2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35055167

ABSTRACT

The receptor tyrosine kinase AXL (RTK-AXL) is implicated in therapy resistance and tumor progression in glioblastoma multiforme (GBM). Here, we investigated therapy-induced receptor modifications and how endogenous RTK-AXL expression and RTK-AXL inhibition contribute to therapy resistance in GBM. GBM cell lines U118MG and SF126 were exposed to temozolomide (TMZ) and radiation (RTX). Receptor modifications in response to therapy were investigated on protein and mRNA levels. TMZ-resistant and RTK-AXL overexpressing cell lines were exposed to increasing doses of TMZ and RTX, with and without RTK-AXL tyrosine kinase inhibitor (TKI). Colorimetric microtiter (MTT) assay and colony formation assay (CFA) were used to assess cell viability. Results showed that the RTK-AXL shedding product, C-terminal AXL (CT-AXL), rises in response to repeated TMZ doses and under hypoxia, acts as a surrogate marker for radio-resistance. Endogenous RTX-AXL overexpression leads to therapy resistance, whereas combination therapy of TZM and RTX with TKI R428 significantly increases therapeutic effects. This data proves the role of RTK-AXL in acquired and intrinsic therapy resistance. By demonstrating that therapy resistance may be overcome by combining AXL TKI with standard treatments, we have provided a rationale for future study designs investigating AXL TKIs in GBM.


Subject(s)
Benzocycloheptenes/pharmacology , Brain Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Glioblastoma/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Temozolomide/pharmacology , Triazoles/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Combined Modality Therapy , Drug Resistance, Neoplasm/radiation effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/metabolism , Glioblastoma/therapy , Humans , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Up-Regulation/drug effects , Up-Regulation/radiation effects , Axl Receptor Tyrosine Kinase
17.
ACS Appl Mater Interfaces ; 14(4): 5112-5121, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35048696

ABSTRACT

Type-I photodynamic therapy (PDT) with less oxygen consumption shows great potential for overcoming the vicious hypoxia typically observed in solid tumors. However, the development of type-I PDT is hindered by insufficient radical generation and the ambiguous design strategy of type-I photosensitizers (PSs). Therefore, developing highly efficient type-I PSs and unveiling their structure-function relationship are still urgent and challenging. Herein, we develop two phenanthro[9,10-d]imidazole derivatives (AQPO and AQPI) with aggregation-induced emission (AIE) characteristics and boost their reactive oxygen species (ROS) generation efficiency by reducing singlet-triplet splitting (ΔEST). Both AQPO and AQPI show ultrasmall ΔEST values of 0.09 and 0.12 eV, respectively. By incorporating electron-rich anisole, the categories of generated ROS by AIE PSs are changed from type-II (singlet oxygen, 1O2) to type-I (superoxide anion radical, O2•- and hydroxyl radical, •OH). We demonstrate that the assembled AQPO nanoparticles (NPs) achieve a 3.2- and 2.9-fold increase in the O2•- and •OH generation efficiencies, respectively, compared to those of AQPI NPs (without anisole) in water, whereas the 1O2 generation efficiency of AQPO NPs is lower (0.4-fold) than that of AQPI NPs. The small ΔEST and anisole group endow AQPO with an excellent capacity for type-I ROS generation. In vitro and in vivo experiments show that AQPO NPs achieve an excellent hypoxia-overcoming PDT effect by efficiently eliminating tumor cells upon white light irradiation with good biosafety.


Subject(s)
Imidazoles/therapeutic use , Neoplasms/drug therapy , Phenanthrolines/therapeutic use , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Tumor Hypoxia/drug effects , A549 Cells , Animals , Drug Carriers/chemistry , Female , Humans , Imidazoles/chemical synthesis , Imidazoles/radiation effects , Light , Mice , Mice, Inbred BALB C , Mice, Nude , NIH 3T3 Cells , Nanoparticles/chemistry , Phenanthrolines/chemical synthesis , Phenanthrolines/radiation effects , Phosphatidylethanolamines/chemistry , Photochemotherapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/radiation effects , Polyethylene Glycols/chemistry
18.
Bioorg Med Chem Lett ; 60: 128583, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35085720

ABSTRACT

Hypoxia imaging agents can play an important role in the tumor treatment by avoiding the worse effect of radiotherapy and chemotherapy due to the tumor hypoxia. Due to the small size and easy coordination, tricarbonyl technetium-99m can be used to label a wide range of imaging agents. In this work, the tricarbonyl 99mTc labeled small-sized hypoxia imaging agents containing 2-nitroimidazoles were prepared, which have different carbon chain lengths between cyclopentadienyl and 2-nitroimidazole, and which have one or two 2-nitroimidazole groups. The results of S180 cell experiment and biodistribution indicated that these molecules have different hypoxic selectivity. When contains one 2-nitroimidazole, as the carbon chain lengthens, which means the molecular volume becomes larger, hypoxia cellular uptake and selectivity decrease in S180 cell uptake experiment. In biodistribution study in mice bearing S180 tumor, Tc-2 (1-cyclopentadienyl-5-(2-nitro-1H-imidazol-1-yl)-pentan-1-one tricarbonyl 99mTc complex), which has intermediate carbon chain, is better due to the more complex factors. Its tumor/blood (T/B) ratio is 3.56 ± 0.25, tumor/muscle(T/M) ratio is 1.73 ± 0.29 and tumor uptake is 2.23 ± 0.24%ID/g at 2 h. Comparing to other tricarbonyl technetium complexes containing one 2-nitroimidazole, the complexes in this work have an advantage in tumor/blood ratio and tumor uptake. This suggests that the small-volume cyclopentadienyl may have an advantage when used as a ligand. When contains two 2-nitroimidazole groups, the complex, 1-cyclopentadienyl-5-di(2-(2-nitro-1H-imidazol-1-yl)ethyl)amino-pentan-1-one tricarbonyl 99mTc complex (Tc-4), has the better results in the cell experiment than those which contain one 2-nitroimidazole group. Thus the hypoxia imaging agent contains two 2-nitroimidazole groups is more advantageous, but further modifications of Tc-4 are needed to improve its clearance rate in the blood, because the increased lipophilicity leads to a decrease in the T/B ratio of Tc-4. In conclusion, small volume hypoxia imaging agents with two 2-nitroimidazole groups may be the trend of development.


Subject(s)
Nitroimidazoles/pharmacology , Organotechnetium Compounds/pharmacology , Radiopharmaceuticals/pharmacology , Tumor Hypoxia/drug effects , Animals , Cell Line, Tumor , Diagnostic Imaging , Dose-Response Relationship, Drug , Mice , Molecular Structure , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Nitroimidazoles/chemical synthesis , Nitroimidazoles/chemistry , Organotechnetium Compounds/chemical synthesis , Organotechnetium Compounds/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Structure-Activity Relationship , Tissue Distribution
19.
ACS Appl Mater Interfaces ; 14(5): 6387-6403, 2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35077131

ABSTRACT

Hypoxia is a well-known feature of malignant solid tumors. To explain the misinterpretation of tumor hypoxia variation during chemotherapy, we developed a DNA origami-based theranostic nanoplatform with an intercalated anticancer anthraquinone as both the chemotherapeutic drug and the photoacoustic contrast agent. The size distribution of the DNA origami nanostructure is 44.5 ± 2.3 nm, whereas the encapsulation efficiency of the drug is 90.7 ± 1.0%, and the drug loading content is 92.2 ± 0.1%. The controlled cumulative release rates were measured in vitro, showing an acidic environment induced rapid drug release. The values of free energy of binding between the drugs and the DNA double helix were calculated through molecular simulations. The cell viability assay was used to characterize cytotoxicity, and fluorescence confocal cell imaging illustrates the biodistribution of the probe in vitro. Photoacoustic and fluorescence imaging were used to indicate drug delivery, release, and biodistribution to predict the drug's chemotherapeutic effect in vivo, whereas the photoacoustic signals were compared with those of deoxygenated/oxygenated hemoglobin to represent the tissue hypoxia/normoxia maps during the chemotherapeutic process and indicate alleviated tumor hypoxia. Staining of tissue sections taken from organs and tumors was used to verify the results of photoacoustic imaging. Our results suggest that photoacoustic imaging can visualize this DNA origami-based theranostic nanoplatform and reveal the mechanisms of chemotherapy on tumor hypoxia.


Subject(s)
Anthraquinones/chemistry , DNA/chemistry , Nanostructures/chemistry , Tumor Hypoxia/drug effects , Animals , Anthraquinones/metabolism , Anthraquinones/pharmacology , Anthraquinones/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Female , Hemolysis/drug effects , Humans , Liver/drug effects , Liver/pathology , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Optical Imaging , Rats , Rats, Sprague-Dawley , Tissue Distribution
20.
Cancer Sci ; 113(3): 916-925, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34931404

ABSTRACT

Metastatic progression of tumors is driven by genetic alterations and tumor-stroma interaction. To elucidate the mechanism underlying the oncogene-induced gastric tumor progression, we have developed an organoid-based model of gastric cancer from GAstric Neoplasia (GAN) mice, which express Wnt1 and the enzymes COX2 and microsomal prostaglandin E synthase 1 in the stomach. Both p53 knockout (GAN-p53KO) organoids and KRASG12V -expressing GAN-p53KO (GAN-KP) organoids were generated by genetic manipulation of GAN mouse-derived tumor (GAN wild-type [WT]) organoids. In contrast with GAN-WT and GAN-p53KO organoids, which manifested Wnt addiction, GAN-KP organoids showed a Wnt-independent phenotype and the ability to proliferate without formation of a Wnt-regulated three-dimensional epithelial architecture. After transplantation in syngeneic mouse stomach, GAN-p53KO cells formed only small tumors, whereas GAN-KP cells gave rise to invasive tumors associated with the development of hypoxia as well as to liver metastasis. Spatial transcriptomics analysis suggested that hypoxia signaling contributes to the metastatic progression of GAN-KP tumors. In particular, such analysis identified a cluster of stromal cells located at the tumor invasive front that expressed genes related to hypoxia signaling, angiogenesis, and cell migration. These cells were also positive for phosphorylated extracellular signal-regulated kinase (ERK), suggesting that mitogen-activated protein kinase (MAPK) signaling promotes development of both tumor and microenvironment. The MEK (MAPK kinase) inhibitor trametinib suppressed the development of GAN-KP gastric tumors, formation of a hypoxic microenvironment, tumor angiogenesis, and liver metastasis. Our findings therefore establish a rationale for application of trametinib to suppress metastatic progression of KRAS-mutated gastric cancer.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Animals , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Neoplasm Metastasis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Tumor Hypoxia/drug effects , Tumor Hypoxia/genetics , Tumor Microenvironment/drug effects , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...