Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Radiat Oncol Biol Phys ; 118(2): 485-497, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37619790

ABSTRACT

PURPOSE: Stress granules (SGs) are cytoplasmic aggregates in which mRNAs and specific proteins are trapped in response to a variety of damaging agents. They participate in the cellular defense mechanisms. Currently, their mechanism of formation in response to ionizing radiation and their role in tumor-cell radiosensitivity remain elusive. METHODS AND MATERIALS: The kinetics of SG formation was investigated after the delivery of photon irradiation at different doses to head and neck squamous cell carcinoma cell lines with different radiosensitivities and the HeLa cervical cancer cell line (used as reference). In parallel, the response to a canonical inducer of SGs, sodium arsenite, was also studied. Immunolabeling of SG-specific proteins and mRNA fluorescence in situ hybridization enabled SG detection and quantification. Furthermore, a ribopuromycylation assay was used to assess the cell translational status. To determine whether reactive oxygen species were involved in SG formation, their scavenging or production was induced by pharmacologic pretreatment in both SCC61 and SQ20B cells. RESULTS: Photon irradiation at different doses led to the formation of cytoplasmic foci that were positive for different SG markers. The presence of SGs gradually increased from 30 minutes to 2 hours postexposure in HeLa, SCC61, and Cal60 radiosensitive cells. In turn, the SQ20B and FaDu radioresistant cells did not form SGs. These results indicated a correlation between sensitivity to photon irradiation and SG formation. Moreover, SG formation was significantly reduced by reactive oxygen species scavenging using dimethyl sulfoxide in SCC61 cells, which supported their role in SG formation. However, a reciprocal experiment in SQ20B cells that depleted glutathione using buthionine sulfoximide did not restore SG formation in these cells. CONCLUSIONS: SGs are formed in response to irradiation in radiosensitive, but not in radioresistant, head and neck squamous cell carcinoma cells. Interestingly, compared with sodium arsenite-induced SGs, photon-induced SGs exhibited a different morphology and cellular localization. Moreover, photon-induced SGs were not associated with the inhibition of translation; rather, they depended on oxidative stress.


Subject(s)
Arsenites , Head and Neck Neoplasms , Sodium Compounds , Stress Granules , Humans , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Reactive Oxygen Species , In Situ Hybridization, Fluorescence , HeLa Cells , Radiation Tolerance , Head and Neck Neoplasms/radiotherapy
2.
Methods Cell Biol ; 180: 69-80, 2023.
Article in English | MEDLINE | ID: mdl-37890933

ABSTRACT

Preclinical development of cancer treatments including radiotherapy (RT) is now crucial to optimize all the treatment aspects for a better efficacy and to help clinicians to build new clinical trials based on robust results. More and more teams use preclinical irradiators to deliver radiotherapy in a comparable way to clinical treatments (image-based RT, arc therapy, stereotactic body RT…). In daily conditions, users usually need to develop easy to use techniques (for applicator technicians for example), allowing to treat many mice per day with a high level of reproducibility. Besides, the best compromise between a satisfying dose coverage to the tumor and nearby organs at risk sparing has to be ensured. We describe here new a home-made immobilization device to irradiate grafted tumors, as well as the different steps to develop the treatment planning and generate an easy procedure to routinely irradiate subcutaneous tumor model.


Subject(s)
Neoplasms , Radiosurgery , Animals , Mice , Reproducibility of Results , Radiotherapy Planning, Computer-Assisted/methods , Neoplasms/radiotherapy , Radiosurgery/methods , Organs at Risk/radiation effects
3.
Int Rev Cell Mol Biol ; 378: 31-60, 2023.
Article in English | MEDLINE | ID: mdl-37438020

ABSTRACT

Natural killer (NK) cells are innate lymphoid cells that play an essential role in the anti-tumor response through immunosurveillance, multiple mechanisms of cytotoxicity and the synthesis of cytokines modulating the immune tumor microenvironment (TME). After the dramatic advances in immunotherapy targeting T cells including the success of checkpoint inhibitors or autologous chimeric antigen receptor (CAR) expressing T cells in clinical practice, NK cells have gained growing interest for the development of new therapies. Although NK cells have shown promising responses in leukemia patients, the effects of NK-targeted therapies are currently limited in the treatment of solid tumors. Thus, radiotherapy could provide a valuable solution to improve treatments targeting NK cells. Indeed, ionizing radiations represent a powerful immuno-modulator that can either induce a pro-inflammatory and anti-tumor TME, or conversely lead to immunosuppression of effector immune cells in favor of tumor growth and therapeutic escape, depending on how it is delivered and tumor models. However, the effects of ionizing radiation on NK cells are only partially understood. Therefore, we review the effects of radiotherapy on the NK cell-mediated anti-tumor response, and propose potential strategies to reinvigorate NK cells by combining radiotherapy with NK cell-targeted therapies.


Subject(s)
Immunity, Innate , Leukemia , Humans , Immunotherapy , Killer Cells, Natural , Cytokines , Tumor Microenvironment
4.
Nucl Med Biol ; 104-105: 53-64, 2022.
Article in English | MEDLINE | ID: mdl-34922279

ABSTRACT

Targeted radionuclide therapy (TRT) is used to treat disseminated or metastatic tumours in which conventional external beam radiotherapy (EBRT) would have unacceptable side effects. Unlike EBRT, TRT delivers low doses at a continuous low dose rate. In EBRT, the effect increases progressively with the dose rate, and biological effects (tumour control and normal tissue damage) are related to the dose according to a sigmoid curve model. This model is part of the so-called quantitative radiobiology that is mostly based on the target cell theory, according to which cell death is due to (lethal) radiation hits to vital cellular targets. This model was developed for EBRT, but was adapted to low dose-rate situations by including a parameter that reflects the time needed to repair tissue damage. However, a growing body of evidence indicates that the model should take into account also the biological effects, which are due to intercellular communications (bystander effects) and amplify the effects of radiation, as well as the immune system. Moreover, extranuclear targets must be considered, although induced intracellular and intercellular signalling pathways may ultimately result in DNA damage. It is likely that bystander effects and immune response always contribute to the overall response to TRT at different levels, and that dose and dose rate are key parameters in controlling their real contribution. We hypothesize that the dose rate is the key determinant in the balance between the physical and DNA-centred response on one side, and the biological response that integrates all subcellular compartments and intercellular signalling pathways on the other side.


Subject(s)
Neoplasms , Radiobiology , DNA Damage , Humans , Neoplasms/radiotherapy , Radioisotopes/therapeutic use , Signal Transduction
5.
Int J Mol Sci ; 21(19)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003449

ABSTRACT

We investigated the potential involvement of ceramide-enriched membrane domains in radiation-induced targeted and nontargeted effects using head and neck squamous cell carcinoma with opposite radiosensitivities. In radiosensitive SCC61 cells, the proportion of targeted effects was 34% and nontargeted effects killed 32% of cells. In contrast, only targeted effects (30%) are involved in the overall death of radioresistant SQ20B cells. We then demonstrated in SCC61 cells that nontargeted cell response was driven by the formation of the radiation-induced ceramide-enriched domain. By contrast, the existence of these platforms in SQ20B cells confers a permissive region for phosphatidylinositol-3-kinase (PI3K)/AKT activation. The disruption of lipid raft results in strong inhibition of PI3K/AKT signaling, leading to radiosensitization and apparition of nontargeted effects. These results suggest that ceramide-enriched platforms play a significant role in targeted and nontargeted effects during radiotherapy and that drugs modulating cholesterol levels may be a good alternative for improving radiotherapy effectiveness.


Subject(s)
Ceramides/pharmacology , Radiation Tolerance/drug effects , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cholesterol/genetics , Combined Modality Therapy , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , Radiation , Signal Transduction/drug effects , Signal Transduction/radiation effects , Squamous Cell Carcinoma of Head and Neck/genetics
6.
Leukemia ; 34(5): 1315-1328, 2020 05.
Article in English | MEDLINE | ID: mdl-31836849

ABSTRACT

Some patients with B-cell non-Hodkin lymphoma Lymphoma (NHL) become refractory to rituximab (anti-CD20 antibody) therapy associated with chemotherapy. Here, the effect of the anti-CD37 antibody-radionuclide conjugate lutetium-177 (177Lu)-lilotomab (Betalutin®) was investigated in preclinical models of NHL. In SCID mice bearing DOHH2 (transformed follicular lymphoma, FL) cell xenografts, 177Lu-lilotomab significantly delayed tumor growth, even at low activity (100 MBq/kg). In athymic mice bearing OCI-Ly8 (diffuse large B-cell lymphoma, DLBCL) or Ramos (Burkitt's lymphoma) cell xenografts, 177Lu-lilotomab activity had to be increased to 500 MBq/kg to show a significant tumor growth delay. Clonogenic and proliferation assays showed that DOHH2 cells were highly sensitive to 177Lu-lilotomab, while Ramos cells were the least sensitive, and U2932 (DLBCL), OCI-Ly8, and Rec-1 (mantle cell lymphoma) cells displayed intermediate sensitivity. The strong 177Lu-lilotomab cytotoxicity observed in DOHH2 cells correlated with reduced G2/M cell cycle arrest, lower WEE-1- and MYT-1-mediated phosphorylation of cyclin-dependent kinase-1 (CDK1), and higher apoptosis. In agreement, 177Lu-lilotomab efficacy in vitro, in vivo, and in patient samples was increased when combined with G2/M cell cycle arrest inhibitors (MK-1775 and PD-166285). These results indicate that 177Lu-lilotomab is particularly efficient in treating tumors with reduced inhibitory CDK1 phosphorylation, such as transformed FL.


Subject(s)
Antibodies, Monoclonal/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , M Phase Cell Cycle Checkpoints/drug effects , Radiopharmaceuticals/pharmacology , Animals , Apoptosis , Cell Proliferation , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 25(15): 4775-4790, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31061069

ABSTRACT

PURPOSE: For the development of new anticancer therapeutic radiopharmaceuticals, including alpha particle emitters, it is important to determine the contribution of targeted effects in irradiated cells, and also of nontargeted effects in nonirradiated neighboring cells, because they may affect the therapeutic efficacy and contribute to side effects. EXPERIMENTAL DESIGN: Here, we investigated the contribution of nontargeted cytotoxic and genotoxic effects in vitro and in vivo (in xenografted mice) during alpha (212Pb/212Bi, 213Bi) and Auger (125I) radioimmunotherapy (RIT). RESULTS: Between 67% and 94% (alpha RIT) and 8% and 15% (Auger RIT) of cancer cells were killed by targeted effects, whereas 7% to 36% (alpha RIT) and 27% to 29% (Auger RIT) of cells were killed by nontargeted effects. We then demonstrated that the nontargeted cell response to alpha and Auger RIT was partly driven by lipid raft-mediated activation of p38 kinase and JNK. Reactive oxygen species also played a significant role in these nontargeted effects, as demonstrated by NF-κB activation and the inhibitory effects of antioxidant enzymes and radical scavengers. Compared with RIT alone, the use of RIT with ASMase inhibitor (imipramine) or with a lipid raft disruptor (e.g., methyl-beta-cyclodextrin or filipin) led to an increase in clonogenic cell survival in vitro and to larger tumors and less tissue DNA damage in vivo. These results were supported by an inhibitory effect of pravastatin on Auger RIT. CONCLUSIONS: Cell membrane-mediated nontargeted effects play a significant role during Auger and alpha RIT, and drugs that modulate cholesterol level, such as statins, could interfere with RIT efficacy.


Subject(s)
Cholesterol/metabolism , Imipramine/pharmacology , MAP Kinase Kinase 4/metabolism , Neoplasms/radiotherapy , Radioimmunotherapy/methods , Radiopharmaceuticals/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bismuth/pharmacology , Cell Line, Tumor , Cell Survival , Female , Filipin/pharmacology , Humans , Iodine Radioisotopes/pharmacology , Lead Radioisotopes/pharmacology , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Radioisotopes/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , beta-Cyclodextrins/pharmacology
8.
J Nucl Med ; 59(8): 1234-1242, 2018 08.
Article in English | MEDLINE | ID: mdl-29674421

ABSTRACT

We have developed the 16F12 mouse monoclonal antibody (mAb), which targets the Müllerian-inhibiting substance receptor, type II (MISRII), expressed by ovarian tumors. Here, we assessed in preclinical models the possibility of using radiolabeled 16F12 in a theranostic approach for small-volume ovarian peritoneal carcinomatosis, such as after cytoreductive surgery. Methods: DOTA-, DTPA- or deferoxamine mesylate-conjugated 16F12 mAb was radiolabeled with ß-particle (177Lu) or α-particle (213Bi) emitters for therapeutic use and with 89Zr for PET imaging. On the 13th postxenograft day, mice bearing intraperitoneal MISRII-positive AN3CA endometrial carcinoma cell xenografts were treated by conventional intraperitoneal radioimmunotherapy (IP-RIT) with 10 MBq of 177Lu-16F12 or 12.9 MBq of 213Bi-16F12 or by brief intraperitoneal radioimmunotherapy (BIP-RIT) using 50 MBq of 177Lu-16F12 or 37 MBq of 213Bi-16F12. For BIP-RIT, 30 min after injection of the radiolabeled mAbs, the peritoneal cavity was washed to remove the unbound radioactivity. The biodistribution of 177Lu- and 213Bi-16F12 mAbs was determined and then used for dose assessment. Hematologic toxicity was also monitored. Results: The 16F12 mAb was satisfactorily radiolabeled for both therapy and imaging. IP-RIT with 177Lu-16F12 was slightly more efficient in delaying tumor growth than IP-RIT with 213Bi-16F12. Conversely, 213Bi-16F12 was more efficient than 177Lu-16F12 in BIP-RIT. The biodistribution analysis showed that the tumor-to-blood uptake ratio was significantly higher with BIP-RIT than with IP-RIT for both 213Bi- and 177Lu-16F12. Hematologic toxicity was more pronounced with 177Lu-16F12 than with 213Bi-16F12. SPECT/CT images (after BIP-RIT with 177Lu-16F12) and PET/CT images (after injection of 89Zr-16F12 in the tail vein) showed focal uptake at the tumor site. Conclusion: Radiolabeled 16F12 could represent a new theranostic tool for small-volume ovarian peritoneal carcinomatosis. Specifically, 213Bi-16F12-based BIP-RIT could be proposed to selected patients as an alternative adjuvant treatment immediately after cytoreductive surgery. An anti-MISRII mAb is currently being used in a first-in-human study, thus making radiolabeled anti-MISRII mAbs a realistic theranostic option for the clinic.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/radiotherapy , Receptors, Peptide/immunology , Receptors, Transforming Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Cell Line, Tumor , Deferoxamine/chemistry , Female , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Isotope Labeling , Mice , Ovarian Neoplasms/metabolism , Pentetic Acid/chemistry , Positron Emission Tomography Computed Tomography , Radiochemistry , Tissue Distribution
9.
Antioxid Redox Signal ; 25(8): 467-84, 2016 09 10.
Article in English | MEDLINE | ID: mdl-27224059

ABSTRACT

AIMS: We investigated whether radiation-induced nontargeted effects are involved in the cytotoxic effects of anticell surface monoclonal antibodies labeled with Auger electron emitters, such as iodine 125 (monoclonal antibodies labeled with (125)I [(125)I-mAbs]). RESULTS: We showed that the cytotoxicity of (125)I-mAbs targeting the cell membrane of p53(+/+) HCT116 colon cancer cells is mainly due to nontargeted effects. Targeted and nontargeted cytotoxicities were inhibited in vitro following lipid raft disruption with Methyl-ß-cyclodextrin (MBCD) or filipin or use of radical oxygen species scavengers. (125)I-mAb efficacy was associated with acid sphingomyelinase activation and modulated through activation of the AKT, extracellular signal-related kinase ½ (ERK1/2), p38 kinase, c-Jun N-terminal kinase (JNK) signaling pathways, and also of phospholipase C-γ (PLC-γ), proline-rich tyrosine kinase 2 (PYK-2), and paxillin, involved in Ca(2+) fluxes. Moreover, the nontargeted response induced by directing 5-[(125)I]iodo-2'-deoxyuridine to the nucleus was comparable to that of (125)I-mAb against cell surface receptors. In vivo, we found that the statistical significance of tumor growth delay induced by (125)I-mAb was removed after MBCD treatment and observed oxidative DNA damage beyond the expected Auger electron range. These results suggest the involvement of nontargeted effects in vivo also. INNOVATION: Low-energy Auger electrons, such as those emitted by (125)I, have a short tissue range and are usually targeted to the nucleus to maximize their cytotoxicity. In this study, we show that targeting the cancer cell surface with (125)I-mAbs produces a lipid raft-mediated nontargeted response that compensates for the inferior efficacy of non-nuclear targeting. CONCLUSION: Our findings describe the mechanisms involved in the efficacy of (125)I-mAbs targeting the cancer cell surface. Antioxid. Redox Signal. 25, 467-484.


Subject(s)
Cell Membrane/metabolism , Cell Membrane/radiation effects , Electrons , Oxidative Stress/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Gene Knockout Techniques , Genes, p53 , HCT116 Cells , Humans , Immunoconjugates/pharmacology , Iodine Radioisotopes/adverse effects , MAP Kinase Signaling System , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Microdomains/radiation effects , Models, Biological , Phosphoproteins/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...