Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Med Phys ; 51(3): 2320-2333, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38345134

ABSTRACT

BACKGROUND: Variable relative biological effectiveness (RBE) models in treatment planning have been proposed to optimize the therapeutic ratio of proton therapy. It has been reported that proton RBE decreases with increasing tumor oxygen level, offering an opportunity to address hypoxia-related radioresistance with RBE-weighted optimization. PURPOSE: Here, we obtain a voxel-level estimation of partial oxygen pressure to weigh RBE values in a single biologically informed beam orientation optimization (BOO) algorithm. METHODS: Three glioblastoma patients with [18 F]-fluoromisonidazole (FMISO)-PET/CT images were selected from the institutional database. Oxygen values were derived from tracer uptake using a nonlinear least squares curve fitting. McNamara RBE, calculated from proton dose, was then weighed using oxygen enhancement ratios (OER) for each voxel and incorporated into the dose fidelity term of the BOO algorithm. The nonlinear optimization problem was solved using a split-Bregman approach, with FISTA as the solver. The proposed hypoxia informed RBE-weighted method (HypRBE) was compared to dose fidelity terms using the constant RBE of 1.1 (cRBE) and the normoxic McNamara RBE model (RegRBE). Tumor homogeneity index (HI), maximum biological dose (Dmax), and D95%, as well as OAR therapeutic index (TI = gEUDCTV /gEUDOAR ) were evaluated along with worst-case statistics after normalization to normal tissue isotoxicity. RESULTS: Compared to [cRBE, RegRBE], HypRBE increased tumor HI, Dmax, and D95% across all plans by on average [31.3%, 31.8%], [48.6%, 27.1%], and [50.4%, 23.8%], respectively. In the worst-case scenario, the parameters increase on average by [12.5%, 14.7%], [7.3%,-8.9%], and [22.3%, 2.1%]. Despite increased OAR Dmean and Dmax by [8.0%, 3.0%] and [13.1%, -0.1%], HypRBE increased average TI by [22.0%, 21.1%]. Worst-case OAR Dmean, Dmax, and TI worsened by [17.9%, 4.3%], [24.5%, -1.2%], and [9.6%, 10.5%], but in the best cases, HypRBE escalates tumor coverage significantly without compromising OAR dose, increasing the therapeutic ratio. CONCLUSIONS: We have developed an optimization algorithm whose dose fidelity term accounts for hypoxia-informed RBE values. We have shown that HypRBE selects bE:\Alok\aaeams better suited to deliver high physical dose to low RBE, hypoxic tumor regions while sparing the radiosensitive normal tissue.


Subject(s)
Glioblastoma , Proton Therapy , Humans , Proton Therapy/methods , Protons , Relative Biological Effectiveness , Positron Emission Tomography Computed Tomography , Radiotherapy Planning, Computer-Assisted/methods , Hypoxia/radiotherapy , Oxygen , Radiotherapy Dosage
2.
J Photochem Photobiol B ; 239: 112643, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36610350

ABSTRACT

Low-level laser therapy, or photobiomodulation, utilizes red or near-infrared light for the treatment of pathological conditions due to the presence of intracellular photoacceptors, such as mitochondrial cytochrome c oxidase, that serve as intermediates for the therapeutic effects. We present an in-detail analysis of the effect of low-intensity LED red light irradiation on the respiratory chain of brain mitochondria. We tested whether low-level laser therapy at 650 nm could alleviate the brain mitochondrial dysfunction in the model of acute hypobaric hypoxia in mice. The irradiation of the mitochondrial fraction of the left cerebral cortex with low-intensity LED red light rescued Complex I-supported respiration during oxidative phosphorylation, normalized the initial polarization of the inner mitochondrial membrane, but has not shown any significant effect on the activity of Complex IV. In comparison, the postponed effect (in 24 h) of the similar transcranial irradiation following hypoxic exposure led to a less pronounced improvement of the mitochondrial functional state, but normalized respiration related to ATP production and membrane polarization. In contrast, the similar irradiation of the mitochondria isolated from control healthy animals exerted an inhibitory effect on CI-supported respiration. The obtained results provide significant insight that can be beneficial for the development of non-invasive phototherapy.


Subject(s)
Brain , Hypoxia , Low-Level Light Therapy , Mitochondria , Animals , Mice , Brain/metabolism , Brain/radiation effects , Electron Transport Complex IV/metabolism , Hypoxia/complications , Hypoxia/metabolism , Hypoxia/radiotherapy , Infrared Rays/therapeutic use , Mitochondria/metabolism , Mitochondria/radiation effects , Pressure/adverse effects , Cell Respiration/radiation effects
3.
Biomed Phys Eng Express ; 8(6)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36260973

ABSTRACT

In proton therapy, a constant relative biological effectiveness (RBE) factor of 1.1 is applied although the RBE has been shown to depend on factors including the Linear Energy Transfer (LET). The biological effectiveness of radiotherapy has also been shown to depend on the level of oxygenation, quantified by the oxygen enhancement ratio (OER). To estimate the biological effectiveness across different levels of oxygenation the RBE-OER-weighted dose (ROWD) can be used. To investigate the consistency between different approaches to estimate ROWD, we implemented and compared OER models in a Monte Carlo (MC) simulation tool. Five OER models were explored: Wenzl and Wilkens 2011 (WEN), Tinganelliet al2015 (TIN), Strigariet al2018 (STR), Dahleet al2020 (DAH) and Meinet al2021 (MEI). OER calculations were combined with a proton RBE model and the microdosimetric kinetic model for ROWD calculations. ROWD and OER were studied for a water phantom scenario and a head and neck cancer case using hypoxia PET data for the OER calculation. The OER and ROWD estimates from the WEN, MEI and DAH showed good agreement while STR and TIN gave higher OER values and lower ROWD. The WEN, STR and DAH showed some degree of OER-LET dependency while this was negligible for the MEI and TIN models. The ROWD for all implemented models is reduced in hypoxic regions with an OER of 1.0-2.1 in the target volume. While some variations between the models were observed, all models display a large difference in the estimated dose from hypoxic and normoxic regions. This shows the potential to increase the dose or LET in hypoxic regions or reduce the dose to normoxic regions which again could lead to normal tissue sparing. With reliable hypoxia imaging, RBE-OER weighting could become a useful tool for proton therapy plan optimization.


Subject(s)
Proton Therapy , Humans , Hypoxia/radiotherapy , Oxygen , Proton Therapy/methods , Radiotherapy Planning, Computer-Assisted/methods , Relative Biological Effectiveness
4.
Int J Radiat Biol ; 98(3): 439-451, 2022.
Article in English | MEDLINE | ID: mdl-34726575

ABSTRACT

PURPOSE: Hypoxia (low oxygen) is a common feature of solid tumors that has been intensely studied for more than six decades. Here we review the importance of hypoxia to radiotherapy with a particular focus on the contribution of hypoxia to immune responses, metastatic potential and FLASH radiotherapy, active areas of research by leading women in the field. CONCLUSION: Although hypoxia-driven metastasis and immunosuppression can negatively impact clinical outcome, understanding these processes can also provide tumor-specific vulnerabilities that may be therapeutically exploited. The different oxygen tensions present in tumors and normal tissues may underpin the beneficial FLASH sparing effect seen in normal tissue and represents a perfect example of advances in the field that can leverage tumor hypoxia to improve future radiotherapy treatments.


Subject(s)
Neoplasms , Radiation Oncology , Female , Humans , Hypoxia/radiotherapy , Immunity , Neoplasms/radiotherapy , Oxygen , Radiotherapy , Radiotherapy Dosage
5.
Sci Rep ; 11(1): 4472, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627727

ABSTRACT

We propose a new in vitro model to assess the impact of 90Y-microspheres derived low-dose beta radiation on colorectal cancer cell line under various oxygenation conditions that mimic the tumor environment. Cancer cells (HCT116) proliferation was assessed using Alamar Blue (AB) assay after 48, 72, and 96 h. FLUKA code assessed changes in cancer cell populations relative to the absorbed dose. In normoxia, mitochondrial activity measured by Alamar Blue after 48-72 h was significantly correlated with the number of microspheres (48 h: r = 0.87 and 72 h: r = 0.89, p < 0.05) and absorbed dose (48 h: r = 0.87 and 72 h: r = 0.7, p < 0.05). In hypoxia, the coefficients were r = 0.43 for both the number of spheres and absorbed dose and r = 0.45, r = 0.47, respectively. Impediment of cancer cell proliferation depended on the absorbed dose. Doses below 70 Gy could reduce colorectal cancer cell proliferation in vitro. Hypoxia induced a higher resistance to radiation than that observed under normoxic conditions. Hypoxia and radiation induced senescence in cultured cells. The new in vitro model is useful for the assessment of 90Y radioembolization effects at the micro-scale.


Subject(s)
Beta Particles/therapeutic use , Colorectal Neoplasms/radiotherapy , Yttrium Radioisotopes/administration & dosage , Cell Proliferation/radiation effects , HCT116 Cells , Humans , Hypoxia/radiotherapy , Microspheres , Mitochondria/radiation effects , Radiometry/methods
6.
Theranostics ; 11(3): 1269-1294, 2021.
Article in English | MEDLINE | ID: mdl-33391534

ABSTRACT

Neonatal hypoxic-ischemic (HI) injury is a severe complication often leading to neonatal death and long-term neurobehavioral deficits in children. Currently, the only treatment option available for neonatal HI injury is therapeutic hypothermia. However, the necessary specialized equipment, possible adverse side effects, and limited effectiveness of this therapy creates an urgent need for the development of new HI treatment methods. Photobiomodulation (PBM) has been shown to be neuroprotective against multiple brain disorders in animal models, as well as limited human studies. However, the effects of PBM treatment on neonatal HI injury remain unclear. Methods: Two-minutes PBM (808 nm continuous wave laser, 8 mW/cm2 on neonatal brain) was applied three times weekly on the abdomen of pregnant rats from gestation day 1 (GD1) to GD21. After neonatal right common carotid artery ligation, cortex- and hippocampus-related behavioral deficits due to HI insult were measured using a battery of behavioral tests. The effects of HI insult and PBM pretreatment on infarct size; synaptic, dendritic, and white matter damage; neuronal degeneration; apoptosis; mitochondrial function; mitochondrial fragmentation; oxidative stress; and gliosis were then assessed. Results: Prenatal PBM treatment significantly improved the survival rate of neonatal rats and decreased infarct size after HI insult. Behavioral tests revealed that prenatal PBM treatment significantly alleviated cortex-related motor deficits and hippocampus-related memory and learning dysfunction. In addition, mitochondrial function and integrity were protected in HI animals treated with PBM. Additional studies revealed that prenatal PBM treatment significantly alleviated HI-induced neuroinflammation, oxidative stress, and myeloid cell/astrocyte activation. Conclusion: Prenatal PBM treatment exerts neuroprotective effects on neonatal HI rats. Underlying mechanisms for this neuroprotection may include preservation of mitochondrial function, reduction of inflammation, and decreased oxidative stress. Our findings support the possible use of PBM treatment in high-risk pregnancies to alleviate or prevent HI-induced brain injury in the perinatal period.


Subject(s)
Hypoxia-Ischemia, Brain/radiotherapy , Hypoxia/radiotherapy , Ischemia/radiotherapy , Animals , Animals, Newborn , Apoptosis/radiation effects , Astrocytes/radiation effects , Cerebral Cortex/radiation effects , Disease Models, Animal , Female , Hippocampus/radiation effects , Low-Level Light Therapy/methods , Male , Mitochondria/radiation effects , Neurons/radiation effects , Neuroprotective Agents/therapeutic use , Oxidative Stress/radiation effects , Pregnancy , Rats , Rats, Sprague-Dawley
7.
Bull Exp Biol Med ; 168(5): 602-604, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32249399

ABSTRACT

The physiological stress modeled by circulatory hypoxia activates LPO processes in various tissues. In posthypoxic period, the infrared low-intensity laser irradiation significantly decreased the chemiluminescence parameters in blood plasma, normalized the retinal levels of diene and triene conjugates, and decreased MDA in the rat brain attesting to the correcting effect of this irradiation during various types of physiological stresses.


Subject(s)
Hypoxia/radiotherapy , Infrared Rays/therapeutic use , Lipid Peroxidation/radiation effects , Phototherapy , Retina/radiation effects , Animals , Antioxidants/metabolism , Antioxidants/radiation effects , Free Radicals/metabolism , Free Radicals/radiation effects , Hypoxia/metabolism , Hypoxia/pathology , Laser Therapy/methods , Lasers , Male , Oxidative Stress/physiology , Oxidative Stress/radiation effects , Phototherapy/methods , Rats , Retina/metabolism
8.
Sci Rep ; 10(1): 1638, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005829

ABSTRACT

Hypoxia, the state of low oxygenation that often arises in solid tumours due to their high metabolism and irregular vasculature, is a major contributor to the resistance of tumours to radiation therapy (RT) and other treatments. Conventional RT extends treatment over several weeks or more, and nominally allows time for oxygen levels to increase ("reoxygenation") as cancer cells are killed by RT, mitigating the impact of hypoxia. Recent advances in RT have led to an increase in the use stereotactic body radiotherapy (SBRT), which delivers high doses in five or fewer fractions. For cancers such as pancreatic adenocarcinoma for which hypoxia varies significantly between patients, SBRT might not be optimal, depending on the extent to which reoxygenation occurs during its short duration. We used fluoro-5-deoxy-α-D-arabinofuranosyl)-2-nitroimidazole positron-emission tomography (FAZA-PET) imaging to quantify hypoxia before and after 5-fraction SBRT delivered to patient-derived pancreatic cancer xenografts orthotopically implanted in mice. An imaging technique using only the pre-treatment FAZA-PET scan and repeat dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) scans throughout treatment was able to predict the change in hypoxia. Our results support the further testing of this technique for imaging of reoxygenation in the clinic.


Subject(s)
Oxygen/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/radiotherapy , Adenocarcinoma/metabolism , Adenocarcinoma/radiotherapy , Animals , Humans , Hypoxia/metabolism , Hypoxia/radiotherapy , Mice , Positron-Emission Tomography/methods , Radiopharmaceuticals/therapeutic use , Radiosurgery/methods , Pancreatic Neoplasms
9.
Cancer Res ; 79(18): 4787-4797, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31311808

ABSTRACT

Hypoxia plays a key role in tumor resistance to radiotherapy. It is important to study hypoxia dynamics during radiotherapy to improve treatment planning and prognosis. Here, we describe a luminescent nanoprobe, composed of a fluorescent semiconducting polymer and palladium complex, for quantitative longitudinal imaging of tumor hypoxia dynamics during radiotherapy. The nanoprobe was designed to provide high sensitivity and reversible response for the subtle change in hypoxia over a narrow range (0-30 mmHg O2), which spans the oxygen range where tumors have limited radiosensitivity. Following intravenous administration, the nanoprobe efficiently accumulated in and distributed across the tumor, including the hypoxic region. The ratio between emissions at 700 and 800 nm provided quantitative mapping of hypoxia across the entire tumor. The nanoprobe was used to image tumor hypoxia dynamics over 7 days during fractionated radiotherapy and revealed that high fractional dose (10 Gy) was more effective in improving tumor reoxygenation than low dose (2 Gy), and the effect tended to persist longer in smaller or more radiosensitive tumors. Our results also indicated the importance of the reoxygenation efficiency of the first fraction in the prediction of the radiation treatment outcome. In summary, this work has established a new nanoprobe for highly sensitive, quantitative, and longitudinal imaging of tumor hypoxia dynamics following radiotherapy, and demonstrated its value for assessing the efficacy of radiotherapy and radiation treatment planning. SIGNIFICANCE: This study presents a novel nanoagent for the visualization and quantification of tumor hypoxia.


Subject(s)
Hypoxia/pathology , Image Processing, Computer-Assisted/methods , Luminescent Agents/chemistry , Molecular Probes/chemistry , Nanoparticles/chemistry , Neoplasms/pathology , Animals , Dose Fractionation, Radiation , Female , Humans , Hypoxia/diagnostic imaging , Hypoxia/radiotherapy , Luminescence , Mice , Mice, Nude , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Spectroscopy, Near-Infrared , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Radiat Oncol ; 14(1): 21, 2019 Jan 29.
Article in English | MEDLINE | ID: mdl-30696472

ABSTRACT

BACKGROUND: Despite the advances in oncology, patients with bulky tumors have worse prognosis and often receive only palliative treatments. Bulky disease represents an important challenging obstacle for all currently available radical treatment options including conventional radiotherapy. The purpose of this study was to assess a retrospective outcome on the use of a newly developed unconventional stereotactic body radiation therapy (SBRT) for PArtial Tumor irradiation of unresectable bulky tumors targeting exclusively their HYpoxic segment (SBRT-PATHY) that exploits the non-targeted effects of radiotherapy: bystander effects (local) and the abscopal effects (distant). MATERIALS AND METHODS: Twenty-three patients with bulky tumors received partial bulky irradiation in order to induce the local non-targeted effect of radiation (bystander effect). The hypoxic tumor segment, called the bystander tumor volume (BTV), was defined using PET and contrast-enhanced CT, as a hypovascularized-hypometabolic junctional zone between the central necrotic and peripheral hypervascularized-hypermetabolic tumor segment. Based on tumor site and volume, the BTV was irradiated with 1-3 fractions of 10-12 Gy prescribed to 70% isodose-line. The pathologic lymph nodes and metastases were not irradiated in order to assess the distant non-targeted effects of radiation (abscopal effect). No patient received any systemic therapy. RESULTS: At the time of analysis, with median follow-up of 9.4 months (range: 4-20), 87% of patients remained progression-free. The bystander and abscopal response rates were 96 and 52%, respectively. Median shrinkage of partially irradiated bulky tumor expressing intensity of the bystander effect was 70% (range 30-100%), whereas for the non-irradiated metastases (intensity of the abscopal effect), it was 50% (range 30-100%). No patient experienced acute or late toxicity of any grade. CONCLUSIONS: SBRT-PATHY showed very inspiring results on exploitation of the radiation-hypoxia-induced non-targeted effects that need to be confirmed through our ongoing prospective trial. Present study has been retrospectively registered by the local ethic committee under study number A 26/18.


Subject(s)
Bystander Effect , Hypoxia/radiotherapy , Neoplasms/pathology , Neoplasms/surgery , Radiosurgery/methods , Radiotherapy Planning, Computer-Assisted/methods , Adult , Aged , Aged, 80 and over , Dose Fractionation, Radiation , Female , Humans , Hypoxia/pathology , Male , Middle Aged , Organs at Risk/radiation effects , Prognosis , Radiotherapy Dosage , Research Design , Retrospective Studies , Tumor Burden
11.
Phys Med Biol ; 64(4): 045008, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30641490

ABSTRACT

We report on a novel method for simultaneous biological optimization of treatment plans for hypoxic tumors using multiple ion species. Our previously introduced kill painting approach, where the overall cell killing is optimized on biologically heterogeneous targets, was expanded with the capability of handling different ion beams simultaneously. The current version (MIBO) of the research treatment planning system TRiP98 has now been augmented to handle 3D (voxel-by-voxel) target oxygenation data. We present a case of idealized geometries where this method can identify optimal combinations leading to an improved peak-to-entrance effective dose ratio. This is achieved by the redistribution of particle fluences, when the heavier ions are preferentially forwarded to hypoxic target areas, while the lighter ions deliver the remaining dose to its normoxic regions. Finally, we present an in silico skull base chordoma patient case study with a combination of 4He and 16O beams, demonstrating specific indications for its potential clinical application. In this particular case, the mean dose, received by the brainstem, was reduced by 3%-5% and by 10%-12% as compared to the pure 4He and 16O plans, respectively. The new method allows a full biological optimization of different ion beams, exploiting the capabilities of actively scanned ion beams of modern particle therapy centers. The possible experimental verification of the present approach at ion beam facilities disposing of fast ion switch is presented and discussed.


Subject(s)
Chordoma/radiotherapy , Helium/therapeutic use , Hypoxia/radiotherapy , Oxygen/therapeutic use , Radiotherapy Planning, Computer-Assisted/methods , Skull Base Neoplasms/radiotherapy , Chordoma/pathology , Humans , Radiotherapy Dosage , Skull Base Neoplasms/pathology
12.
Antioxid Redox Signal ; 28(2): 131-140, 2018 Jan 10.
Article in English | MEDLINE | ID: mdl-28741367

ABSTRACT

AIMS: Evofosfamide (TH-302) is a hypoxia-activated prodrug (HAP) that releases the DNA-damaging bromo-isophosphoramide mustard (Br-IPM) moiety selectively under hypoxic conditions. Since solid tumors are known to have hypoxic regions, HAPs in combination with chemotherapy or radiotherapy (XRT) will be beneficial. We tested the oxygen dependence of release kinetics of Br-IPM using electron paramagnetic resonance (EPR) with spin trapping by monitoring redox cycling of the nitroimidazole moiety of TH-302, and oxygen dependence of TH-302 on in vitro cytotoxicity at different levels of hypoxia was also examined. Two tumor implants (SCCVII and HT29) in mice were studied. RESULTS: TH-302 fragmentation to release Br-IPM was noticed at oxygen levels <76 mmHg, which increased with higher levels of hypoxia. Enhanced cellular cytotoxicity was also observed at oxygen levels <76 mmHg. In vivo pO2 imaging in the two tumor implants showed that the SCCVII tumor implant had higher level of hypoxia compared with the HT29 xenograft. TH-302 as a monotherapy in vivo showed modest effects in SCCVII implants and minimal effects in HT29 xenografts, whereas TH-302 in combination with ionizing radiation showed significant benefit in both tumor models. INNOVATION: We examined the kinetics of redox cycling versus fragmentation of TH-302. The combination of oxygen-dependent XRT with TH-302 is effective even in tumors with significant hypoxia. CONCLUSIONS: Imaging studies identifying the magnitude of hypoxia in tumors indicated that the responsiveness to TH-302 and the antitumor effect of TH-302 were enhanced by combining with XRT in both the TH-302-sensitive SCCVII tumor and -resistant HT29 tumor. Antioxid. Redox Signal. 28, 131-140.


Subject(s)
Hypoxia/metabolism , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Prodrugs , Radiotherapy , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Disease Models, Animal , Humans , Hypoxia/drug therapy , Hypoxia/radiotherapy , Mice , Oxidation-Reduction/drug effects , Oxidation-Reduction/radiation effects , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
13.
Food Funct ; 8(8): 2857-2864, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28726915

ABSTRACT

Radiotherapy in ovarian cancer frequently invokes resistance; this severely compromises its therapeutic effect and results in poor clinical prognosis. How to overcome the acquired resistance and re-sensitize tumors to radiation is the central question in this clinical setting. Cancer cell survival was evaluated using a clonogenic assay. The microRNA expression profile was analyzed using a microarray. Transcript expression was determined using real time PCR. The expression of protein was determined by immunoblotting. Transcription activation was measured using a luciferase reporter assay. Transcription factor binding was determined using ChIP-PCR. Xenograft model was established and exposed to radiation with the simultaneous administration of oleuropein. Tumor growth was monitored. We demonstrated that treatment of oleuropein-sensitized ovarian cells with radiation altered the microRNA expression profile. The endogenous expression of miR-299 was suppressed by a hypoxia inducible factor and relieved in response to oleuropein, which in turn suppressed HPSE1 expression and consequently led to increased sensitivity to radiation. Our data elucidates an unappreciated mechanism mediating radiotherapy resistance in ovarian cancer and exploits the potential synergistic effect of oleuropein with radiation, which warrants further clinical investigation.


Subject(s)
Glucuronidase/genetics , Hypoxia/radiotherapy , Iridoids/administration & dosage , MicroRNAs/genetics , Ovarian Neoplasms/radiotherapy , Radiation-Sensitizing Agents/administration & dosage , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glucuronidase/antagonists & inhibitors , Glucuronidase/metabolism , Humans , Hypoxia/drug therapy , Hypoxia/enzymology , Hypoxia/genetics , Iridoid Glucosides , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics
14.
Acta Oncol ; 56(6): 819-825, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28464740

ABSTRACT

BACKGROUND: Hypoxia imaged by positron emission tomography (PET) is a potential target for optimization in radiotherapy. However, the implementation of this approach with respect to the conversion of intensities in the images into oxygenation and radiosensitivity maps is not straightforward. This study investigated the feasibility of applying two conversion approaches previously derived for 18F-labeled fluoromisonidazole (18F-FMISO)-PET images for the hypoxia tracer 18F-flortanidazole (18F-HX4). MATERIAL AND METHODS: Ten non-small-cell lung cancer patients imaged with 18F-HX4 before the start of radiotherapy were considered in this study. PET image uptake was normalized to a well-oxygenated reference region and subsequently linear and non-linear conversions were used to determine tissue oxygenations maps. These were subsequently used to delineate hypoxic volumes based partial oxygen pressure (pO2) thresholds. The results were compared to hypoxic volumes segmented using a tissue-to-background ratio of 1.4 for 18F-HX4 uptake. RESULTS: While the linear conversion function was not found to result in realistic oxygenation maps, the non-linear function resulted in reasonably sized sub-volumes in good agreement with uptake-based segmented volumes for a limited range of pO2 thresholds. However, the pO2 values corresponding to this range were significantly higher than what is normally considered as hypoxia. The similarity in size, shape, and relative location between uptake-based sub-volumes and volumes based on the conversion to pO2 suggests that the relationship between uptake and pO2 is similar for 18F-FMISO and 18F-HX4, but that the model parameters need to be adjusted for the latter. CONCLUSIONS: A non-linear conversion function between uptake and oxygen partial pressure for 18F-FMISO-PET could be applied to 18F-HX4 images to delineate hypoxic sub-volumes of similar size, shape, and relative location as based directly on the uptake. In order to apply the model for e.g., dose-painting, new parameters need to be derived for the accurate calculation of dose-modifying factors for this tracer.


Subject(s)
Aorta/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Hypoxia/pathology , Lung Neoplasms/pathology , Muscles/pathology , Positron-Emission Tomography/methods , Radiotherapy, Image-Guided/methods , Aorta/diagnostic imaging , Aorta/radiation effects , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/radiotherapy , Fluorodeoxyglucose F18 , Humans , Hypoxia/diagnostic imaging , Hypoxia/radiotherapy , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/radiotherapy , Muscles/diagnostic imaging , Muscles/radiation effects , Radiopharmaceuticals , Radiotherapy Dosage , Reference Standards
15.
Int J Hyperthermia ; 33(7): 770-778, 2017 11.
Article in English | MEDLINE | ID: mdl-28540811

ABSTRACT

The gadolinium-doped iron oxide nanoparticles (GdIONP) with greater specific power adsorption rate (SAR) than Fe3O4 was developed and its potential application in tumour therapy and particle tracking were demonstrated in transgenic adenocarcinoma of the mouse prostate C1 (TRAMP-C1) tumours. The GdIONPs accumulated in tumour region during the treatment could be clearly tracked and quantified by T2-weighted MR imaging. The therapeutic effects of GdIONP-mediated hyperthermia alone or in combination with radiotherapy (RT) were also evaluated. A significant increase in the tumour growth time was observed following the treatment of thermotherapy (TT) only group (2.5 days), radiation therapy only group (4.5 days), and the combined radio-thermotherapy group (10 days). Immunohistochemical staining revealed a reduced hypoxia region with vascular disruption and extensive tumour necrosis following the combined radio-thermotherapy. These results indicate that GdIONP-mediated hyperthermia can improve the efficacy of RT by its dual functions in high temperature (temperature greater than 45 °C)-mediated thermal ablation and mild-temperature hyperthermia (MTH) (temperature between 39 and 42 °C)-mediated reoxygenation.


Subject(s)
Gadolinium/therapeutic use , Hyperthermia, Induced , Magnetic Fields , Magnetite Nanoparticles/therapeutic use , Prostatic Neoplasms/therapy , Animals , Cell Line, Tumor , Combined Modality Therapy , Hypoxia/pathology , Hypoxia/radiotherapy , Hypoxia/therapy , Male , Mice, Inbred C57BL , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Tumor Burden
16.
Oncotarget ; 7(47): 76613-76627, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27780936

ABSTRACT

The extent of tumor oxygenation is an important factor contributing to the efficacy of radiation therapy (RTx). Interestingly, several preclinical studies have shown benefit of combining RTx with drugs that inhibit tumor blood vessel growth, i.e. angiostatic therapy. Recent findings show that proper scheduling of both treatment modalities allows dose reduction of angiostatic drugs without affecting therapeutic efficacy. We found that whilst low dose sunitinib (20 mg/kg/day) did not affect the growth of xenograft HT29 colon carcinoma tumors in nude mice, the combination with either single dose RTx (1x 5Gy) or fractionated RTx (5x 2Gy/week, up to 3 weeks) substantially hampered tumor growth compared to either RTx treatment alone. To better understand the interaction between RTx and low dose angiostatic therapy, we explored the effects of RTx on tumor angiogenesis and tissue perfusion. DCE-MRI analyses revealed that fractionated RTx resulted in enhanced perfusion after two weeks of treatment. This mainly occurred in the center of the tumor and was accompanied by increased tissue viability and decreased hypoxia. These effects were accompanied by increased expression of the pro-angiogenic growth factors VEGF and PlGF. DCE-MRI and contrast enhanced ultrasonography showed that the increase in perfusion and tissue viability was counteracted by low-dose sunitinib. Overall, these data give insight in the dynamics of tumor perfusion during conventional 2 Gy fractionated RTx and provide a rationale to combine low dose angiostatic drugs with RTx both in the palliative as well as in the curative setting.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Neoplasms/pathology , Neovascularization, Pathologic , Radiotherapy , Animals , Cell Line, Tumor , Chemoradiotherapy , Combined Modality Therapy , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Hypoxia/drug therapy , Hypoxia/metabolism , Hypoxia/radiotherapy , Magnetic Resonance Imaging/methods , Mice , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms/therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/radiotherapy , Radiotherapy/methods , Ultrasonography/methods
17.
Cell Prolif ; 49(3): 304-14, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27079860

ABSTRACT

OBJECTIVES: Advanced head and neck carcinomas (HNCs) are aggressive tumours, mainly due to hypoxia and a cancer stem cell (CSC) subpopulation. The aim of this study was to simulate tumour growth and behaviour during radiotherapy of three HNC groups (governed by different growth kinetics, hypoxia levels and CSC division pattern) to determine correlation between resistance factors and responses to hyperfractionated radiotherapy. METHODS: An in silico HNC model was developed based on biologically realistic input parameters. During radiotherapy simulation, three parameters were studied: growth kinetics, hypoxia and probability of CSC symmetrical division. Both independent and combined effects on tumour response to hyperfractionated radiotherapy were assessed. RESULTS: Oxic and very mildly hypoxic HNCs were revealed to be controlled by hyperfractionated radiotherapy, irrespective of growth kinetics and CSC division pattern. Moderately hypoxic tumours had different responses to radiotherapy: while slowly proliferating HNCs were still controllable, tumours with higher cell turnover were more resistant. In rapidly proliferating tumours, the number of fractions needed for tumour control increased exponentially with the probability of CSC symmetrical division, whereas in moderately growing HNC, this behaviour was linear. Severely hypoxic tumours could not be controlled by radiotherapy alone. Tumours with CSCs in a severely hypoxic niche required adjuvant therapies to be eradicated. CONCLUSIONS: Growth kinetics strongly influence tumour responses to treatment. Slowly growing tumours showed linear dependence between dose and hypoxia/CSC, whereas rapidly growing tumours followed exponential behaviour.


Subject(s)
Head and Neck Neoplasms/radiotherapy , Hypoxia/radiotherapy , Neoplastic Stem Cells/radiation effects , Cell Proliferation , Computer Simulation , Head and Neck Neoplasms/complications , Head and Neck Neoplasms/pathology , Humans , Hypoxia/complications , Hypoxia/pathology , Models, Biological , Monte Carlo Method , Neoplastic Stem Cells/pathology
18.
Int J Hyperthermia ; 31(6): 693-701, 2015.
Article in English | MEDLINE | ID: mdl-25986432

ABSTRACT

PURPOSE: The tumour microenvironment is frequently hypoxic, poorly perfused, and exhibits abnormally high interstitial fluid pressure. These factors can significantly reduce efficacy of chemo and radiation therapies. The present study aims to determine whether mild systemic heating alters these parameters and improves response to radiation in human head and neck tumour xenografts in SCID mice. MATERIALS AND METHODS: SCID mice were injected with FaDu cells (a human head and neck carcinoma cell line), or implanted with a resected patient head and neck squamous cell carcinoma grown as a xenograft, followed by mild systemic heating. Body temperature during heating was maintained at 39.5 ± 0.5 °C for 4 h. Interstitial fluid pressure (IFP), hypoxia and relative tumour perfusion in the tumours were measured at 2 and 24 h post-heating. Tumour vessel perfusion was measured 24 h post-heating, coinciding with the first dose of fractionated radiotherapy. RESULTS: Heating tumour-bearing mice resulted in significant decrease in intratumoural IFP, increased the number of perfused tumour blood vessels as well as relative tumour perfusion in both tumour models. Intratumoural hypoxia was also reduced in tumours of mice that received heat treatment. Mice bearing FaDu tumours heated 24 h prior to five daily radiation treatments exhibited significantly enhanced tumour response compared to tumours in control mice. CONCLUSIONS: Mild systemic heating can significantly alter the tumour microenvironment of human head and neck tumour xenograft models, decreasing IFP and hypoxia while increasing microvascular perfusion. Collectively, these effects could be responsible for the improved response to radiotherapy.


Subject(s)
Head and Neck Neoplasms , Hyperthermia, Induced , Animals , Cell Line, Tumor , Extracellular Fluid , Female , Fluorescent Dyes/administration & dosage , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , Head and Neck Neoplasms/therapy , Humans , Hypoxia/pathology , Hypoxia/radiotherapy , Hypoxia/therapy , Liposomes , Mice, SCID , Pilot Projects , Pressure , Transplantation, Heterologous , Tumor Burden , Tumor Microenvironment
19.
Asian Pac J Cancer Prev ; 16(2): 627-33, 2015.
Article in English | MEDLINE | ID: mdl-25684498

ABSTRACT

BACKGROUND: To study the effect of parecoxib, a novel cyclooxygenase-2 selective inhibitor, on the radiation response of colorectal cancer (CRC) cells and its underlying mechanisms. MATERIALS AND METHODS: Both in vitro colony formation and apoptosis assays as well as in vivo mouse xenograft experiments were used to explore the radiosensitizing effects of parecoxib in human HCT116 and HT29 CRC cells. RESULTS: Parecoxib sensitized CRC cells to radiation in vitro with a sensitivity enhancement ratio of 1.32 for HCT116 cells and 1.15 for HT29 cells at a surviving fraction of 0.37. This effect was partially attributable to enhanced apoptosis induction by parecoxib combined with radiation, as illustrated using an in vitro apoptosis assays. Parecoxib augmented the tumor response of HCT116 xenografts to radiation, achieving growth delay more than 20 days and an enhancement factor of 1.53. In accordance with the in vitro results, parecoxib combined with radiation resulted in less proliferation and more apoptosis in tumors than radiation alone. Radiation monotherapy decreased microvessel density (MVD) and microvessel intensity (MVI), but increased the hypoxia level in xenografts. Parecoxib did not affect MVD, but it increased MVI and attenuated hypoxia. CONCLUSIONS: Parecoxib can effectively enhance radiation sensitivity in CRC cells through direct effects on tumor cells and indirect effects on tumor vasculature.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Cyclooxygenase 2 Inhibitors/pharmacology , Hypoxia/drug therapy , Isoxazoles/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/radiation effects , Blotting, Western , Cell Proliferation/radiation effects , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Flow Cytometry , Humans , Hypoxia/pathology , Hypoxia/radiotherapy , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Tumor Cells, Cultured , X-Rays , Xenograft Model Antitumor Assays
20.
J R Soc Interface ; 12(103)2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25540239

ABSTRACT

Glioblastoma multiforme (GBM) is a highly invasive primary brain tumour that has poor prognosis despite aggressive treatment. A hallmark of these tumours is diffuse invasion into the surrounding brain, necessitating a multi-modal treatment approach, including surgery, radiation and chemotherapy. We have previously demonstrated the ability of our model to predict radiographic response immediately following radiation therapy in individual GBM patients using a simplified geometry of the brain and theoretical radiation dose. Using only two pre-treatment magnetic resonance imaging scans, we calculate net rates of proliferation and invasion as well as radiation sensitivity for a patient's disease. Here, we present the application of our clinically targeted modelling approach to a single glioblastoma patient as a demonstration of our method. We apply our model in the full three-dimensional architecture of the brain to quantify the effects of regional resistance to radiation owing to hypoxia in vivo determined by [(18)F]-fluoromisonidazole positron emission tomography (FMISO-PET) and the patient-specific three-dimensional radiation treatment plan. Incorporation of hypoxia into our model with FMISO-PET increases the model-data agreement by an order of magnitude. This improvement was robust to our definition of hypoxia or the degree of radiation resistance quantified with the FMISO-PET image and our computational model, respectively. This work demonstrates a useful application of patient-specific modelling in personalized medicine and how mathematical modelling has the potential to unify multi-modality imaging and radiation treatment planning.


Subject(s)
Brain Neoplasms , Glioma , Hypoxia , Misonidazole/analogs & derivatives , Models, Biological , Positron-Emission Tomography , Radiation-Sensitizing Agents/administration & dosage , Aged , Brain Neoplasms/blood supply , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/radiotherapy , Glioma/blood supply , Glioma/diagnostic imaging , Glioma/radiotherapy , Humans , Hypoxia/diagnostic imaging , Hypoxia/radiotherapy , Male , Misonidazole/administration & dosage , Precision Medicine , Radiography
SELECTION OF CITATIONS
SEARCH DETAIL
...