Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 363
1.
Int J Mol Sci ; 25(8)2024 Apr 19.
Article En | MEDLINE | ID: mdl-38674080

Cellular hypoxia, detectable in up to 80% of non-small cell lung carcinoma (NSCLC) tumors, is a known cause of radioresistance. High linear energy transfer (LET) particle radiation might be effective in the treatment of hypoxic solid tumors, including NSCLC. Cellular hypoxia can activate nuclear factor κB (NF-κB), which can modulate radioresistance by influencing cancer cell survival. The effect of high-LET radiation on NF-κB activation in hypoxic NSCLC cells is unclear. Therefore, we compared the effect of low (X-rays)- and high (12C)-LET radiation on NF-κB responsive genes' upregulation, as well as its target cytokines' synthesis in normoxic and hypoxic A549 NSCLC cells. The cells were incubated under normoxia (20% O2) or hypoxia (1% O2) for 48 h, followed by irradiation with 8 Gy X-rays or 12C ions, maintaining the oxygen conditions until fixation or lysis. Regulation of NF-κB responsive genes was evaluated by mRNA sequencing. Secretion of NF-κB target cytokines, IL-6 and IL-8, was quantified by ELISA. A greater fold change increase in expression of NF-κB target genes in A549 cells following exposure to 12C ions compared to X-rays was observed, regardless of oxygenation status. These genes regulate cell migration, cell cycle, and cell survival. A greater number of NF-κB target genes was activated under hypoxia, regardless of irradiation status. These genes regulate cell migration, survival, proliferation, and inflammation. X-ray exposure under hypoxia additionally upregulated NF-κB target genes modulating immunosurveillance and epithelial-mesenchymal transition (EMT). Increased IL-6 and IL-8 secretion under hypoxia confirmed NF-κB-mediated expression of pro-inflammatory genes. Therefore, radiotherapy, particularly with X-rays, may increase tumor invasiveness in surviving hypoxic A549 cells.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , NF-kappa B , Humans , NF-kappa B/metabolism , A549 Cells , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Lung Neoplasms/pathology , Lung Neoplasms/genetics , X-Rays , Gene Expression Regulation, Neoplastic/radiation effects , Linear Energy Transfer , Cell Hypoxia/radiation effects , Carbon , Cell Survival/radiation effects , Radiation Tolerance , Interleukin-8/metabolism , Interleukin-8/genetics
2.
FASEB J ; 36(5): e22331, 2022 05.
Article En | MEDLINE | ID: mdl-35476363

The blood-brain barrier (BBB) regulates molecular and cellular entry from the cerebrovasculature into the surrounding brain parenchyma. Many diseases of the brain are associated with dysfunction of the BBB, where hypoxia is a common stressor. However, the contribution of hypoxia to BBB dysfunction is challenging to study due to the complexity of the brain microenvironment. In this study, we used a BBB model with brain microvascular endothelial cells and pericytes differentiated from iPSCs to investigate the effect of hypoxia on barrier function. We found that hypoxia-induced barrier dysfunction is dependent upon increased actomyosin contractility and is associated with increased fibronectin fibrillogenesis. We propose a role for actomyosin contractility in mediating hypoxia-induced barrier dysfunction through modulation of junctional claudin-5. Our findings suggest pericytes may protect brain microvascular endothelial cells from hypoxic stresses and that pericyte-derived factors could be candidates for treatment of pathological barrier-forming tissues.


Actomyosin , Blood-Brain Barrier , Claudin-5 , Endothelial Cells , Pericytes , Actomyosin/metabolism , Blood-Brain Barrier/metabolism , Cell Hypoxia/radiation effects , Claudin-5/metabolism , Culture Media, Conditioned , Endothelial Cells/metabolism , Humans , Pericytes/metabolism
3.
J Nanobiotechnology ; 19(1): 315, 2021 Oct 12.
Article En | MEDLINE | ID: mdl-34641905

BACKGROUND: Sonodynamic therapy (SDT) strategies exhibit a high tissue penetration depth and can achieve therapeutic efficacy by facilitating the intertumoral release of reactive oxygen species (ROS) with a short lifespan and limited diffusion capabilities. The majority of SDT systems developed to date are of the highly O2-dependent type II variety, limiting their therapeutic utility in pancreatic cancer and other hypoxic solid tumor types. RESULTS: Herein, a nucleus-targeted ultra-small Ti-tetrakis(4-carboxyphenyl)porphyrin (TCPP) metal-organic framework (MOF) platform was synthesized and shown to be an effective mediator of SDT. This MOF was capable of generating large quantities of ROS in an oxygen-independent manner in response to low-intensity ultrasound (US) irradiation (0.5 W cm-2), thereby facilitating both type I and type II SDT. This approach thus holds great promise for the treatment of highly hypoxic orthotopic pancreatic carcinoma solid tumors. This Ti-TCPP MOF was able to induce in vitro cellular apoptosis by directly destroying DNA and inducing S phase cell cycle arrest following US irradiation. The prolonged circulation, high intratumoral accumulation, and nucleus-targeting attributes of these MOF preparations significantly also served to significantly inhibit orthotopic pancreatic tumor growth and prolong the survival of tumor-bearing mice following Ti-TCPP + US treatment. Moreover, this Ti-TCPP MOF was almost completely cleared from mice within 7 days of treatment, and no apparent treatment-associated toxicity was observed. CONCLUSION: The nucleus-targeted ultra-small Ti-TCPP MOF developed herein represents an effective approach to the enhanced SDT treatment of tumors in response to low-intensity US irradiation.


Metal-Organic Frameworks/pharmacology , Pancreatic Neoplasms/therapy , Ultrasonic Therapy , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neoplasms, Experimental/therapy , Pancreatic Neoplasms
4.
Nat Rev Clin Oncol ; 18(12): 751-772, 2021 12.
Article En | MEDLINE | ID: mdl-34326502

Hypoxia is prevalent in human tumours and contributes to microenvironments that shape cancer evolution and adversely affect therapeutic outcomes. Historically, two different tumour microenvironment (TME) research communities have been discernible. One has focused on physicochemical gradients of oxygen, pH and nutrients in the tumour interstitium, motivated in part by the barrier that hypoxia poses to effective radiotherapy. The other has focused on cellular interactions involving tumour and non-tumour cells within the TME. Over the past decade, strong links have been established between these two themes, providing new insights into fundamental aspects of tumour biology and presenting new strategies for addressing the effects of hypoxia and other microenvironmental features that arise from the inefficient microvascular system in solid tumours. This Review provides a perspective on advances at the interface between these two aspects of the TME, with a focus on translational therapeutic opportunities relating to the elimination and/or exploitation of tumour hypoxia.


Neoplasms/therapy , Oxygen/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Humans , Neoplasms/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects
5.
Nanotechnology ; 32(42)2021 Jul 27.
Article En | MEDLINE | ID: mdl-34319255

Photodynamic therapy (PDT) and immunotherapy have been often adopted for ovarian cancer therapy, yet their application is limited by the high recurrence rate and toxic side effects. Intriguingly, nanoparticles contribute to enhancing the performance of PDT. Here, we investigated the synthesis of HER-2-Nanobody (Nb)-conjugated human serum albumin (HSA) incorporated with chlorin (Ce6) and catalase (CAT) (Nb@HCC), and analyzed the synergic effect of Nb@HCC-mediated PDT and immunotherapy for SK-OV-3 tumors. The Ce6 and CAT were incorporated into HSA to construct the HCC nanoparticles. HER-2-Nanobody was the purified bacterial crude extract, and conjugated with HCC to prepare Nb@HCC via heterodisulfide. The effects of Nb@HCC with near infrared ray (NIR) irradiation on moderating hypoxia and hypoxia inducible factor-1α(HIF-1α) expression were evaluated in the SK-OV-3 cells and tumor tissues. A SK-OV-3 tumor-bearing model was developed, where the synergistic effect of Nb@HCC-mediated PDT and anti-CTLA-4 therapy was investigated. Nb@HCC with a 660 nm laser irradiation could induce massive reactive oxygen species and trigger apoptosis in SK-OV-3 cells. Nb@HCC and PDT promoted danger-associated molecular patterns (DAMPs), which indicated immunogenic cell death and maturation of dendritic cells in the SK-OV-3 cells. Irradiated by NIR, Nb@HCC alleviated the hypoxia and decreased the expression of HIF-1α. The Nb@HCC-mediated PDT and anti-CTLA-4 therapy synergically inhibited the progression of distant tumor, and induced T cell infiltration. Biosafety tests suggested that Nb@HCC would not cause damage to the major organs with less toxicity and side effects. To conclude, a combination of Nb@HCC-mediated PDT and anti-CTLA-4 therapy could inhibit the progression of distant tumor to attain remarkable therapeutic outcomes.


Cell Hypoxia , Ovarian Neoplasms/metabolism , Photochemotherapy/methods , Single-Domain Antibodies , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Catalase , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Porphyrins , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/toxicity
6.
Br J Cancer ; 125(1): 85-93, 2021 07.
Article En | MEDLINE | ID: mdl-33846523

BACKGROUND: miRNAs are promising biomarkers in oncology as their small size makes them less susceptible to degradation than mRNA in FFPE tissue. We aimed to derive a hypoxia-associated miRNA signature for bladder cancer. METHODS: Taqman miRNA array cards identified miRNA seed genes induced under hypoxia in bladder cancer cell lines. A signature was derived using feature selection methods in a TCGA BLCA training data set. miRNA expression data were generated for 190 tumours from the BCON Phase 3 trial and used for independent validation. RESULTS: A 14-miRNA hypoxia signature was derived, which was prognostic for poorer overall survival in the TCGA BLCA cohort (n = 403, p = 0.001). Univariable analysis showed that the miRNA signature predicted an overall survival benefit from having carbogen-nicotinamide with radiotherapy (HR = 0.30, 95% CI 0.094-0.95, p = 0.030) and performed similarly to a 24-gene mRNA signature (HR = 0.47, 95% CI 0.24-0.92, p = 0.025). Combining the signatures improved performance (HR = 0.26, 95% CI 0.08-0.82, p = 0.014) with borderline significance for an interaction test (p = 0.065). The interaction test was significant for local relapse-free survival LRFS (p = 0.033). CONCLUSION: A 14-miRNA hypoxia signature can be used with an mRNA hypoxia signature to identify bladder cancer patients benefitting most from having carbogen and nicotinamide with radiotherapy.


Carbon Dioxide/administration & dosage , MicroRNAs/genetics , Niacinamide/administration & dosage , Oxygen/administration & dosage , Urinary Bladder Neoplasms/therapy , Biomarkers, Tumor/genetics , Carbon Dioxide/pharmacology , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Chemoradiotherapy , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Niacinamide/pharmacology , Oligonucleotide Array Sequence Analysis , Oxygen/pharmacology , Prognosis , Survival Analysis , Urinary Bladder Neoplasms/genetics
7.
Biomed Res Int ; 2021: 8880179, 2021.
Article En | MEDLINE | ID: mdl-33532500

Regulated necrosis (necroptosis) is crucially involved in cardiac ischaemia-reperfusion injury (MIRI). The aim of our study is to investigate whether shock wave therapy (SWT) is capable of exerting protective effects by inhibiting necroptosis during myocardial ischaemia-reperfusion (I/R) injury and the possible role of autophagy in this process. We established a hypoxia/reoxygenation (H/R) model in vitro using HL-1 cells to simulate MIRI. MTS assays and LDH cytotoxicity assay were performed to measure cell viability and cell damage. Annexin V/PI staining was used to determine apoptosis and necrosis. Western blotting was performed to assess the changes in cell signaling pathways associated with autophagy, necroptosis, and apoptosis. Reactive oxygen species (ROS) production was detected using DHE staining. Autophagosome generation and degradation (autophagic flux) were analysed using GFP and RFP tandemly tagged LC3 (tfLC3). HL-1 cells were then transfected with p62/SQSTM1 siRNA in order to analyse its role in cardioprotection. Our results revealed that SWT increased cell viability in the H/R model and decreased receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 expression. ROS production was also inhibited by SWT. Moreover, SWT decreased Beclin1 expression and the ratio of LC3-II/LC3-I following H/R. Simultaneously, in the tfLC3 assay, the SWT provoked a decrease in the cumulative autophagosome abundance. siRNA-mediated knockdown of p62 attenuated H/R-induced necroptosis, and SWT did not exert additive effects. Taken together, SWT ameliorated H/R injury by inhibiting necroptosis. SWT also relieved the blockade of autophagic flux in response to H/R injury. The restoration of autophagic flux by SWT might contribute to its cardioprotective effect on necroptosis following H/R injury.


Autophagy/radiation effects , Cell Hypoxia/radiation effects , Extracorporeal Shockwave Therapy , Myocytes, Cardiac , Necroptosis/radiation effects , Animals , Cell Line , Cell Survival/radiation effects , Heart/radiation effects , Mice , Models, Biological , Myocardial Reperfusion Injury , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/radiation effects
8.
Curr Radiopharm ; 14(1): 46-50, 2021.
Article En | MEDLINE | ID: mdl-32228432

BACKGROUND: Tumors are defined as abnormal tissue masses, and one of the most important factors leading to the growth of these abnormal tissue masses is Vascular Endothelial Growth Factor, which stimulates angiogenesis by releasing cells under hypoxic conditions. Hypoxia has a vital role in cancer therapy, thus it is important to monitor hypoxia. The hypoxia marker Pimonidazole (PIM) is a candidate biomarker of cancer aggressiveness. OBJECTIVE: The study aimed to perform radioiodination of PIM with Iodine-131 (131I) to join a theranostic approach. For this purpose, PIM was derived as PIM-TOS to be able to be radioiodinated. METHODS: PIM was derived via a tosylation reaction. Derivatization product (PIM-TOS) was radioiodinated by using iodogen method and was analyzed by High-Performance Liquid Chromatography and Liquid chromatography-mass spectrometry. Thin layer radiochromatography was utilized for its quality control studies. RESULTS: PIM was derived successfully after the tosylation reaction. The radioiodination yield of PIM-TOS was over 85%. CONCLUSION: In the current study, radioiodination potential of PIM with 131I, as a potential theranostic hypoxia agent was investigated. Further experimental studies should be performed for developing a novel hypoxia probe including theranostics approaches.


Cell Hypoxia/radiation effects , Iodine Radioisotopes/administration & dosage , Nitroimidazoles/radiation effects , Precision Medicine/methods , Radiation-Sensitizing Agents/radiation effects , Radiopharmaceuticals/administration & dosage , Humans
9.
Mol Med Rep ; 23(1)2021 01.
Article En | MEDLINE | ID: mdl-33215223

The current study investigated whether hyperoxia may reverse hypoxia­induced radioresistance (RR) in cervical cancer. Human HeLa cells exposed to hypoxic, normoxic or hyperoxic conditions were irradiated using X­rays. Cell proliferation and apoptosis were analyzed using MTT assays and flow cytometry. The expression levels of hypoxia­inducible factor­1α (HIF­1α), VEGF165, VEGFRs, Akt and ERK were measured via western blotting and/or ELISA. The results demonstrated that hypoxia stimulated HIF­1α and VEGF expression, and induced RR in HeLa cells. The administration of recombinant VEGF or the forced expression of VEGF promoted RR, whereas inactivating HIF­1α or blocking the VEGF­VEGFR interaction abrogated hypoxia­induced RR. Notably, hyperoxia decreased the level of hypoxia­stimulated HIF­1α and VEGF, and enhanced radiosensitivity in hypoxic HeLa cells. The results demonstrated that hyperoxia suppressed the hypoxia­activated Akt and ERK signaling pathways in HeLa cells. Therefore, a high O2 concentration may be considered as a radiotherapeutic sensitizer for hypoxic HeLa cells.


Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oxygen/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Apoptosis , Cell Hypoxia/radiation effects , Cell Proliferation , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Int J Mol Sci ; 21(22)2020 Nov 10.
Article En | MEDLINE | ID: mdl-33182844

Hypofractionation is currently considered a valid alternative to conventional radiotherapy for the treatment of patients with organ-confined prostate cancer. Recent data have demonstrated that extreme hypofractionation, which involves the use of a high radiation dose per delivered fraction and concomitant reduction of sessions, is a safe and effective treatment, even though its radiobiological rationale is still lacking. The present work aims to investigate the biological basis sustaining this approach and to evaluate the potential of a hypofractionated regimen in combination with androgen deprivation therapy, one of the major standards of care for prostate cancer. Findings show that androgen receptor (AR) modulation, by use of androgens and antiandrogens, has a significant impact on cell survival, especially in hypoxic conditions (4% O2). Subsequent experiments have revealed that AR activity as a transcription factor is involved in the onset of malignant senescence-associated secretory phenotype (SASP) and activation of DNA repair cascade. In particular, we found that AR stimulation in hypoxic conditions promotes the enhanced transcription of ATM gene, the cornerstone kinase of the DNA damage repair genes. Together, these data provide new potential insights to justify the use of androgen deprivation therapy, in particular with second-generation anti-androgens such as enzalutamide, in combination with radiotherapy.


Androgen Antagonists/therapeutic use , Chemoradiotherapy/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Androgen Receptor Antagonists/therapeutic use , Androgens/therapeutic use , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , DNA Repair/genetics , Humans , Male , Metribolone/pharmacology , Models, Biological , Prostatic Neoplasms/metabolism , Radiation Dose Hypofractionation , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcriptome
11.
Int J Mol Sci ; 21(10)2020 May 14.
Article En | MEDLINE | ID: mdl-32423018

The demand for personalized medicine in radiotherapy has been met by a surge of mechanistic models offering predictions of the biological effect of ionizing radiation under consideration of a growing number of parameters. We present an extension of our existing model of cell survival after photon irradiation to explicitly differentiate between the damage inflicted by the direct and indirect (radicals-mediated) action of ionizing radiation. Within our approach, we assume that the oxygenation status affects the indirect action. The effect of different concentrations of dimethyl sulfoxide (DMSO), an effective radical scavenger, has been simulated at different dose levels in normoxic and hypoxic conditions for various cell lines. Our model is found to accurately predict experimental data available in literature, validating the assumptions made in our approach. The presented extension adds further flexibility to our model and could act as basis for further developments of our model.


Cell Hypoxia/radiation effects , Cell Survival/radiation effects , DNA Damage/radiation effects , Dimethyl Sulfoxide/pharmacology , Cell Hypoxia/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Radiation , Free Radical Scavengers/pharmacology , Humans , Photons/adverse effects
12.
Int J Mol Sci ; 21(9)2020 May 08.
Article En | MEDLINE | ID: mdl-32397263

Photodynamic therapy (PDT) has been used to treat certain types of non-melanoma skin cancer with promising results. However, some skin lesions have not fully responded to this treatment, suggesting a potential PDT-resistant phenotype. Therefore, novel therapeutic alternatives must be identified that improve PDT in resistant skin cancer. In this study, we analyzed the cell viability, intracellular protoporphyrin IX (PpIX) content and subcellular localization, proliferation profile, cell death, reactive oxygen species (ROS) detection and relative gene expression in PDT-resistant HSC-1 cells. PDT-resistant HSC-1 cells show a low quantity of protoporphyrin IX and low levels of ROS, and thus a low rate of death cell. Furthermore, the resistant phenotype showed a downregulation of HSPB1, SLC15A2, FECH, SOD2 and an upregulation of HMBS and BIRC5 genes. On the other hand, epigallocatechin gallate catechin enhanced the MAL-PDT effect, increasing levels of protoporphyrin IX and ROS, and killing 100% of resistant cells. The resistant MAL-PDT model of skin cancer squamous cells (HSC-1) is a reliable and useful tool to understand PDT cytotoxicity and cellular response. These resistant cells were successfully sensitized with epigallocatechin gallate catechin. The in vitro epigallocatechin gallate catechin effect as an enhancer of MAL-PDT in resistant cells is promising in the treatment of difficult skin cancer lesions.


Anticarcinogenic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Catechin/analogs & derivatives , Cell Death/drug effects , Cell Proliferation/drug effects , Combined Modality Therapy/methods , Photochemotherapy/methods , Skin Neoplasms/drug therapy , Aminolevulinic Acid/analogs & derivatives , Aminolevulinic Acid/pharmacology , Carcinoma, Squamous Cell/radiotherapy , Catechin/pharmacology , Cell Death/radiation effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Ferrochelatase/genetics , Ferrochelatase/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Photosensitizing Agents/metabolism , Protoporphyrins/metabolism , Reactive Oxygen Species/metabolism , Skin Neoplasms/radiotherapy , Stress, Physiological/drug effects , Stress, Physiological/genetics , Stress, Physiological/radiation effects , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Survivin/genetics , Survivin/metabolism , Symporters/genetics , Symporters/metabolism
13.
Nanoscale ; 12(6): 4051-4060, 2020 Feb 14.
Article En | MEDLINE | ID: mdl-32022048

Hypoxia, one of the features of most solid tumors, can severely impede the efficiency of oxygen-dependent treatments such as chemotherapy, radiotherapy and type-II photodynamic therapy. Herein, a catalase-like nanozyme RuO2@BSA (RB) was first prepared through a biomineralization strategy, and a high efficiency near-infrared photosensitizer (IR-808-Br2) was further loaded into the protein shell to generate the safe and versatile RuO2@BSA@IR-808-Br2 (RBIR) for the imaging-guided enhanced phototherapy against hypoxic tumors. RB not only acts like a catalase, but also serves as a photothermal agent that speeds up the oxygen supply under near-infrared irradiation (808 nm). The loaded NIR photosensitizer could immediately convert molecular oxygen (O2) to cytotoxic singlet oxygen (1O2) upon the same laser irradiation. Results indicated that RBIR achieved enhanced therapeutic outcomes with negligible side effects. Features such as a simple synthetic route and imaging-guided and single-wavelength-excited phototherapy make the nanozyme a promising agent for clinical applications.


Antineoplastic Agents , Cell Hypoxia , Low-Level Light Therapy/methods , Photochemotherapy/instrumentation , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biomineralization , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Mice , Nanostructures/chemistry , Neoplasms, Experimental
14.
Pathol Oncol Res ; 26(1): 433-442, 2020 Jan.
Article En | MEDLINE | ID: mdl-30406875

Radiation Therapy (RT) is a treatment option for a large number of neoplasias. However, the effect of RT on the level of hypoxia markers is poorly understood. The present study aimed to investigate the effect of RT on the levels of hypoxic markers in Oral squamous cell carcinoma (OSCC). Evaluation of HIF-1α and miR-210 levels in OSCC was performed. Then a proteomic analysis was performed to identify candidate hypoxic targets of RT. To validate proteomic studies, the effect of RT on HIF-1α, miR-210, PDH-A and LDH-A levels under hypoxia was assessed by qRT-PCR. The impact of RT in hypoxia markers was evaluated in patients to confirm in vitro results. An increase in the HIF-1α levels was observed in OSCC. RT reduced OSCC cell proliferation and migration. Interestingly, hypoxia could revert the effect of radiation on OSCC phenotype. However, proteomics analyses suggested that LDH is one of the critical targets of RT even in hypoxia. Moreover, RT decreased HIF-1α, miR-210, and LDH even in hypoxia. The current study demonstrated that hypoxia could revert the effects of RT in the OSCC context. However, RT reduces the levels HIF-1α, miR-210 and LDH in vivo and in vitro. The consequences of RT in blood should be carefully investigated.


Cell Hypoxia/radiation effects , Hypoxia-Inducible Factor 1, alpha Subunit/radiation effects , L-Lactate Dehydrogenase/radiation effects , MicroRNAs/radiation effects , Radiotherapy/adverse effects , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/blood , L-Lactate Dehydrogenase/blood , Male , MicroRNAs/blood , Middle Aged , Radiation Tolerance , Young Adult
15.
Cancer Res ; 80(2): 139-150, 2020 01 15.
Article En | MEDLINE | ID: mdl-31767626

The ataxia-telangiectasia mutated (ATM) protein kinase is widely known for its function as a chief mobilizer of the DNA damage response (DDR) upon DNA double-strand breaks. ATM orchestrates the DDR by modulating the expression of various miRNAs through several mechanisms. On the other hand, a set of miRNAs contribute to tight regulation of ATM by directly targeting the 3'-untranslated region of ATM mRNA. This review addresses the therapeutic application and molecular mechanisms that underlie the intricate interactions between miRNAs and ATM. It also describes therapeutic delivery of miRNAs in different environments such as hypoxic tumor microenvironments.


Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Chemoradiotherapy/methods , MicroRNAs/metabolism , Neoplasms/therapy , Radiation Tolerance/genetics , 3' Untranslated Regions/genetics , Antagomirs/pharmacology , Antagomirs/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Hypoxia/genetics , Cell Hypoxia/radiation effects , Combined Modality Therapy/methods , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Neoplasms/genetics , Radiation Tolerance/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Tumor Microenvironment/genetics , Tumor Microenvironment/radiation effects
16.
Cancer Res ; 79(23): 6044-6053, 2019 12 01.
Article En | MEDLINE | ID: mdl-31641030

There is increasing evidence that high doses of radiotherapy, like those delivered in stereotactic body radiotherapy (SBRT), trigger indirect mechanisms of cell death. Such effect seems to be two-fold. High doses may trigger an immune response and may cause vascular damage, leading to cell starvation and death. Development of mathematical response models, including indirect death, may help clinicians to design SBRT optimal schedules. Despite increasing experimental literature on indirect tumor cell death caused by vascular damage, efforts on modeling this effect have been limited. In this work, we present a biomathematical model of this effect. In our model, tumor oxygenation is obtained by solving the reaction-diffusion equation; radiotherapy kills tumor cells according to the linear-quadratic model, and also endothelial cells (EC), which can trigger loss of functionality of capillaries. Capillary death will affect tumor oxygenation, driving nearby tumor cells into severe hypoxia. Capillaries can recover functionality due to EC proliferation. Tumor cells entering a predetermined severe hypoxia status die according to a hypoxia-death model. This model fits recently published experimental data showing the effect of vascular damage on surviving fractions. It fits surviving fraction curves and qualitatively reproduces experimental values of percentages of functional capillaries 48 hours postirradiation, and hypoxic cells pre- and 48 hours postirradiation. This model is useful for exploring aspects of tumor and EC response to radiotherapy and constitutes a stepping stone toward modeling indirect tumor cell death caused by vascular damage and accounting for this effect during SBRT planning. SIGNIFICANCE: A novel biomathematical model of indirect tumor cell death caused by vascular radiation damage could potentially help clinicians interpret experimental data and design better radiotherapy schedules.


Apoptosis/radiation effects , Endothelium, Vascular/radiation effects , Models, Biological , Neoplasms/radiotherapy , Radiosurgery/methods , Capillaries/cytology , Capillaries/pathology , Capillaries/radiation effects , Cell Hypoxia/radiation effects , Cell Proliferation/radiation effects , Dose-Response Relationship, Radiation , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Humans , Neoplasms/blood supply , Neoplasms/pathology , Radiotherapy Planning, Computer-Assisted/methods , Treatment Outcome
17.
Medicine (Baltimore) ; 98(40): e17067, 2019 Oct.
Article En | MEDLINE | ID: mdl-31577699

Hypoxia is a well-recognized biological characteristic to therapy resistance and negative prognostic factor in patients with head and neck squamous cell carcinoma (HNSCC). This study aims to investigate the changes of hypoxia measured by F-fluoroerythronitroimidazole (FETNIM) uptake on integrated positron emission tomography and computed tomography (PET/CT) during chemoradiotherapy and its prognostic value of clinical outcome in locoregionally advanced HNSCC.Thirty-two patients with locoregionally advanced HNSCC who received definitive treatment with concurrent chemoradiotherapy underwent FETNIM PET/CT scans before and after 5 weeks of treatment. The intensity of hypoxia using the maximum standardized uptake value (SUVmax) was evaluated both on primary lesion and metastatic lymph node (MLN). The pre-SUVmax and mid-SUVmax were defined as SUVmax on pre- and mid-FETNIM PET/CT. The local control (LC), regional control (RC), distant metastatic-free survival (DMFS), and overall survival (OS) were collected in patient follow-ups.Mid-SUVmax decreased significantly both in the primary tumor (t = 8.083, P < .001) and MLN (t = 6.808, P < .001) compared to pre-SUVmax. With a median follow-up of 54 months, the 5-year LC, RC, DMFS, and OS rates were 55%, 66.7%, 64.7%, and 55%, respectively, for all of the patients. On univariate analysis, patients with high pre-SUVmax in primary tumor had significantly worse LC (56.3% vs 87.5%, P = .046) and OS (43.8% vs 87.5%, P = .023) than other patients. Patients with high mid-SUVmax had significantly worse DMFS (50% vs 84.6%, P = .049) and OS (33.3% vs 73.1%, P = .028) than other patients. The tumor grade and mid-SUVmax were the significant predictors of OS on multivariate analysis.In this study, hypoxia in tumor significantly decreased during chemoradiotherapy. The persistent hypoxia predicted poor OS. The data provided evidence that FETNIM PET/CT could be used dynamically for selecting appropriate patients and optimal timing of hypoxia-adapted therapeutic regimens.


Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Chemoradiotherapy/adverse effects , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/therapy , Positron Emission Tomography Computed Tomography/methods , Adult , Aged , Aged, 80 and over , Chemoradiotherapy/methods , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Nitroimidazoles/administration & dosage , Radiopharmaceuticals/administration & dosage , Survival Analysis , Young Adult
18.
Undersea Hyperb Med ; 46(4): 385-397, 2019.
Article En | MEDLINE | ID: mdl-31509895

Over the past four decades, hyperbaric oxygen (HBO2) therapy has played a prominent role in both the prevention and treatment of mandibular osteoradionecrosis (ORN). It has done so on the strength of laboratory observations and clinical reports, yet only limited efficacy data. This dual role has come under increasing scrutiny in the modern radiotherapy (RT) and surgical eras. The ability to spare healthy "non-target" tissue has markedly improved since the two-dimensional planning and delivery techniques in use when HBO2's prophylactic value was first demonstrated. A recent study failed to identify this same benefit in patients who received high-precision imaging and conformal RT. HBO2 therapy is under challenge as preferred treatment for early stage ORN. A recently introduced "fibroatrophic" mechanism contrasts with the hypovascular-hypocellular-hypoxic injury pattern that formed the basis for HBO2's therapeutic use. This alternative pathophysiologic state appears to benefit from an oral antioxidant medication regimen. The continuing necessity of HBO2 in support of mandibular reconstruction for advanced ORN is in question. Microsurgery-based vascularized bone flaps increasingly represent standard care, invariably in the absence of perioperative HBO2. Renewed interest in hyperbaric oxygen as a radiation sensitizer offers some promise. Hypoxia remains a critical radio-resistant factor in many solid tumors. Malignant gliomas have been a primary focus of several small studies, with resulting improvements in local control and median survival. Hyperbaric radiation sensitization has recently addressed oropharyngeal cancer. Preliminary data indicates that addition of HBO2 to chemo-radiation standard of care is technically feasible, well tolerated and safe. A Phase II efficacy trial will investigate the potential for of HBO2 to improve progression-free and relapse-free survival in newly diagnosed locally advanced head and neck cancers. What follows is a review and summary of relevant peer-reviewed literature.


Head and Neck Neoplasms/radiotherapy , Hyperbaric Oxygenation , Mandible/radiation effects , Osteoradionecrosis/therapy , Radiation Tolerance , Cell Hypoxia/radiation effects , Clinical Trials, Phase II as Topic , Clodronic Acid/therapeutic use , Drug Combinations , Humans , Mandible/surgery , Osteoradionecrosis/pathology , Osteoradionecrosis/prevention & control , Pentoxifylline/therapeutic use , Radiotherapy, Conformal/adverse effects , Plastic Surgery Procedures , Tocopherols/therapeutic use , Tooth Extraction
19.
Acta Oncol ; 58(10): 1476-1482, 2019 Oct.
Article En | MEDLINE | ID: mdl-31432722

Introduction: Positron emission tomography (PET) using hypoxia-selective tracers like FAZA may guide radiation dose-escalation approaches. However, poor resolution combined with slow tracer retention in relatively inaccessible target cells and slow clearance of unbound tracer results in low-contrast images, and areas where viable hypoxic tracer retaining cells and necrosis (no tracer) are intermixed may pass unnoticed during image thresholding. Here we hypothesized that a clinical feasible one-day dual tracer approach that combines a short-lived (e.g., 11C labeled) metabolic tracer that provides voxel-wise information on viable tissue volume (preferably independently of tumor microenvironment) and a hypoxia marker, may limit threshold-based errors. Material and methods: 11C-acetate and 11C-methionine uptake was quantified in tumor cell lines under tumor microenvironment-mimicking conditions of high/low O2 (21%/0%) and pH (7.4/6.7). Next, tumor-bearing mice were administered FAZA and sacrificed 1 h (mimics a clinical low-contrast image scenario) or 4 h (high contrast) later. In addition, all mice were administered pimonidazole (hypoxia) and 14C-methionine 1 h prior to sacrifice. Tumor tissue sections were analyzed using dual-tracer autoradiography. Finally, FAZA, or FAZA normalized to 14C-methionine retention (to adjust for differences in viable tissue volume) was compared to hypoxic fraction (deduced from immune-histological analysis of pimonidazole; ground truth) in PET-mimicking macroscopic pixels with variable extent of necrosis/hypoxia. Results/conclusions: Low pH stimulated 11C-acetate retention in many cell lines, and uptake was further modified by anoxia, compromising its usefulness as a universal marker of viable tumor volume. In contrast, 11C-methionine was largely unaffected by the in vitro microenvironment and was further tested in mice. Necrosis increased the risk of missing hypoxia-containing pixels during thresholding and hypoxic fraction and FAZA signal correlated poorly in the low contrast-scenario. Voxel-based normalization to 14C-methionine increased the likelihood of detecting voxels harboring hypoxic cells profoundly, but did not consistently improve the correlation between the density of hypoxic cells and tracer signal.


Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Radiation Tolerance , Radiopharmaceuticals/administration & dosage , Tumor Burden/radiation effects , Animals , Autoradiography/methods , Cell Hypoxia/radiation effects , Cell Line, Tumor , Female , Humans , Male , Mice , Necrosis/diagnostic imaging , Neoplasms/pathology , Neoplasms/radiotherapy , Nitroimidazoles/administration & dosage , Tumor Microenvironment/radiation effects , Xenograft Model Antitumor Assays
20.
Radiat Res ; 192(3): 258-266, 2019 09.
Article En | MEDLINE | ID: mdl-31265788

Exposure of the gastrointestinal (GI) tract to ionizing radiation can cause acute and delayed injury. However, critical cellular targets that regulate the development of radiation-induced GI injury remain incompletely understood. Here, we investigated the role of vascular endothelial cells in controlling acute and delayed GI injury after total-abdominal irradiation (TAI). To address this, we used genetically engineered mice in which endothelial cells are sensitized to radiation due to the deletion of the tumor suppressor p53. Remarkably, we found that VE-cadherin-Cre; p53FL/FL mice, in which both alleles of p53 are deleted in endothelial cells, were not sensitized to the acute GI radiation syndrome, but these mice were highly susceptible to delayed radiation enteropathy. Histological examination indicated that VE-cadherin-Cre; p53FL/FL mice that developed delayed radiation enteropathy had severe vascular injury in the small intestine, which was manifested by hemorrhage, loss of microvessels and tissue hypoxia. In addition, using dual-energy CT imaging, we showed that VE-cadherin-Cre; p53FL/FL mice had a significant increase in vascular permeability of the small intestine in vivo 28 days after TAI. Together, these findings demonstrate that while sensitization of endothelial cells to radiation does not exacerbate the acute GI radiation syndrome, it is sufficient to promote the development of late radiation enteropathy.


Endothelial Cells/pathology , Endothelial Cells/radiation effects , Intestines/pathology , Intestines/radiation effects , Radiation Tolerance , Animals , Capillary Permeability/radiation effects , Cell Hypoxia/radiation effects , Endothelial Cells/metabolism , Gene Deletion , Mice , Time Factors , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
...