Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Int J Radiat Oncol Biol Phys ; 120(1): 287-300, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38493902

RÉSUMÉ

PURPOSE: We conducted a multi-institutional dosimetric audit between FLASH and conventional dose rate (CONV) electron irradiations by using an anatomically realistic 3-dimensional (3D) printed mouse phantom. METHODS AND MATERIALS: A computed tomography (CT) scan of a live mouse was used to create a 3D model of bony anatomy, lungs, and soft tissue. A dual-nozzle 3D printer was used to print the mouse phantom using acrylonitrile butadiene styrene (∼1.02 g/cm3) and polylactic acid (∼1.24 g/cm3) simultaneously to simulate soft tissue and bone densities, respectively. The lungs were printed separately using lightweight polylactic acid (∼0.64 g/cm3). Hounsfield units (HU), densities, and print-to-print stability of the phantoms were assessed. Three institutions were each provided a phantom and each institution performed 2 replicates of irradiations at selected anatomic regions. The average dose difference between FLASH and CONV dose distributions and deviation from the prescribed dose were measured with radiochromic film. RESULTS: Compared with the reference CT scan, CT scans of the phantom demonstrated mass density differences of 0.10 g/cm3 for bone, 0.12 g/cm3 for lung, and 0.03 g/cm3 for soft tissue regions. Differences in HU between phantoms were <10 HU for soft tissue and bone, with lung showing the most variation (54 HU), but with minimal effect on dose distribution (<0.5%). Mean differences between FLASH and CONV decreased from the first to the second replicate (4.3%-1.2%), and differences from the prescribed dose decreased for both CONV (3.6%-2.5%) and FLASH (6.4%-2.7%). Total dose accuracy suggests consistent pulse dose and pulse number, although these were not specifically assessed. Positioning variability was observed, likely due to the absence of robust positioning aids or image guidance. CONCLUSIONS: This study marks the first dosimetric audit for FLASH using a nonhomogeneous phantom, challenging conventional calibration practices reliant on homogeneous phantoms. The comparison protocol offers a framework for credentialing multi-institutional studies in FLASH preclinical research to enhance reproducibility of biologic findings.


Sujet(s)
Poumon , Fantômes en imagerie , Impression tridimensionnelle , Tomodensitométrie , Animaux , Souris , Poumon/effets des radiations , Poumon/imagerie diagnostique , Radiométrie/méthodes , Dosimétrie en radiothérapie , Polyesters , Électrons , Os et tissu osseux/imagerie diagnostique , Os et tissu osseux/effets des radiations , Polystyrènes , Résines acryliques , Butadiènes
2.
Int J Radiat Oncol Biol Phys ; 119(3): 1001-1010, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38171387

RÉSUMÉ

PURPOSE: Ultrahigh-dose-rate (FLASH) irradiation has been reported to reduce normal tissue damage compared with conventional dose rate (CONV) irradiation without compromising tumor control. This proof-of-concept study aims to develop a deep learning (DL) approach to quantify the FLASH isoeffective dose (dose of CONV that would be required to produce the same effect as the given physical FLASH dose) with postirradiation mouse intestinal histology images. METHODS AND MATERIALS: Eighty-four healthy C57BL/6J female mice underwent 16 MeV electron CONV (0.12 Gy/s; n = 41) or FLASH (200 Gy/s; n = 43) single fraction whole abdominal irradiation. Physical dose ranged from 12 to 16 Gy for FLASH and 11 to 15 Gy for CONV in 1 Gy increments. Four days after irradiation, 9 jejunum cross-sections from each mouse were hematoxylin and eosin stained and digitized for histological analysis. CONV data set was randomly split into training (n = 33) and testing (n = 8) data sets. ResNet101-based DL models were retrained using the CONV training data set to estimate the dose based on histological features. The classical manual crypt counting (CC) approach was implemented for model comparison. Cross-section-wise mean squared error was computed to evaluate the dose estimation accuracy of both approaches. The validated DL model was applied to the FLASH data set to map the physical FLASH dose into the isoeffective dose. RESULTS: The DL model achieved a cross-section-wise mean squared error of 0.20 Gy2 on the CONV testing data set compared with 0.40 Gy2 of the CC approach. Isoeffective doses estimated by the DL model for FLASH doses of 12, 13, 14, 15, and 16 Gy were 12.19 ± 0.46, 12.54 ± 0.37, 12.69 ± 0.26, 12.84 ± 0.26, and 13.03 ± 0.28 Gy, respectively. CONCLUSIONS: Our proposed DL model achieved accurate CONV dose estimation. The DL model results indicate that in the physical dose range of 13 to 16 Gy, the biologic dose response of small intestinal tissue to FLASH irradiation is represented by a lower isoeffective dose compared with the physical dose. Our DL approach can be a tool for studying isoeffective doses of other radiation dose modifying interventions.


Sujet(s)
Apprentissage profond , Souris de lignée C57BL , Animaux , Souris , Femelle , Intestins/effets des radiations , Intestins/anatomopathologie , Dosimétrie en radiothérapie , Jéjunum/effets des radiations , Jéjunum/anatomopathologie , Étude de validation de principe
3.
Integr Biol (Camb) ; 152023 04 11.
Article de Anglais | MEDLINE | ID: mdl-37874173

RÉSUMÉ

Radiation therapy, one of the most effective therapies to treat cancer, is highly toxic to healthy tissue. The delivery of radiation at ultra-high dose rates, FLASH radiation therapy (FLASH), has been shown to maintain therapeutic anti-tumor efficacy while sparing normal tissues compared to conventional dose rate irradiation (CONV). Though promising, these studies have been limited mainly to murine models. Here, we leveraged enteroids, three-dimensional cell clusters that mimic the intestine, to study human-specific tissue response to radiation. We observed enteroids have a greater colony growth potential following FLASH compared with CONV. In addition, the enteroids that reformed following FLASH more frequently exhibited proper intestinal polarity. While we did not observe differences in enteroid damage across groups, we did see distinct transcriptomic changes. Specifically, the FLASH enteroids upregulated the expression of genes associated with the WNT-family, cell-cell adhesion, and hypoxia response. These studies validate human enteroids as a model to investigate FLASH and provide further evidence supporting clinical study of this therapy. Insight Box Promising work has been done to demonstrate the potential of ultra-high dose rate radiation (FLASH) to ablate cancerous tissue, while preserving healthy tissue. While encouraging, these findings have been primarily observed using pre-clinical murine and traditional two-dimensional cell culture. This study validates the use of human enteroids as a tool to investigate human-specific tissue response to FLASH. Specifically, the work described demonstrates the ability of enteroids to recapitulate previous in vivo findings, while also providing a lens through which to probe cellular and molecular-level responses to FLASH. The human enteroids described herein offer a powerful model that can be used to probe the underlying mechanisms of FLASH in future studies.


Sujet(s)
Techniques de culture cellulaire , Intestins , Humains , Souris , Animaux
4.
ArXiv ; 2023 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-37808098

RÉSUMÉ

We conducted a multi-institutional audit of dosimetric variability between FLASH and conventional dose rate (CONV) electron irradiations by using an anatomically realistic 3D-printed mouse phantom. A CT scan of a live mouse was used to create a 3D model of bony anatomy, lungs, and soft tissue. A dual-nozzle 3D printer was used to print the mouse phantom using acrylonitrile butadiene styrene ($~1.02 g/cm^3$) and polylactic acid ($~1.24 g/cm^3$) simultaneously to simulate soft tissue and bone densities, respectively. The lungs were printed separately using lightweight polylactic acid ($~0.64 g/cm^3$). Hounsfield units (HU) and densities were compared with the reference CT scan of the live mouse. Print-to-print reproducibility of the phantom was assessed. Three institutions were each provided a phantom, and each institution performed two replicates of irradiations at selected mouse anatomic regions. The average dose difference between FLASH and CONV dose distributions and deviation from the prescribed dose were measured with radiochromic film. Compared to the reference CT scan, CT scans of the phantom demonstrated mass density differences of $0.10 g/cm^3$ for bone, $0.12 g/cm^3$ for lung, and $0.03 g/cm^3$ for soft tissue regions. Between phantoms, the difference in HU for soft tissue and bone was <10 HU from print to print. Lung exhibited the most variation (54 HU) but minimally affected dose distribution (<0.5% dose differences between phantoms). The mean difference between FLASH and CONV from the first replicate to the second decreased from 4.3% to 1.2%, and the mean difference from the prescribed dose decreased from 3.6% to 2.5% for CONV and 6.4% to 2.7% for FLASH. The framework presented here is promising for credentialing of multi-institutional studies of FLASH preclinical research to maximize the reproducibility of biological findings.

5.
Front Immunol ; 12: 746469, 2021.
Article de Anglais | MEDLINE | ID: mdl-34777358

RÉSUMÉ

Classical dendritic cells (cDCs) in mice have been divided into 2 major subsets based on the expression of nuclear transcription factors: a CD8+Irf8+Batf3 dependent (DC1) subset, and a CD8-Irf4+ (DC2) subset. We found that the CD8+DC1 subset can be further divided into CD8+DC1a and CD8+DC1b subsets by differences in surface receptors, gene expression, and function. Whereas all 3 DC subsets can act alone to induce potent Th1 cytokine responses to class I and II MHC restricted peptides derived from ovalbumin (OVA) by OT-I and OT-II transgenic T cells, only the DC1b subset could effectively present glycolipid antigens to natural killer T (NKT) cells. Vaccination with OVA protein pulsed DC1b and DC2 cells were more effective in reducing the growth of the B16-OVA melanoma as compared to pulsed DC1a cells in wild type mice. In conclusion, the Batf3-/- dependent DC1 cells can be further divided into two subsets with different immune functional profiles in vitro and in vivo.


Sujet(s)
Antigènes CD8/immunologie , Cellules dendritiques/immunologie , Activation des lymphocytes/immunologie , Transcriptome/immunologie , Animaux , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Phénotype
6.
Blood Adv ; 5(17): 3290-3302, 2021 09 14.
Article de Anglais | MEDLINE | ID: mdl-34432869

RÉSUMÉ

Replacement of failed organs followed by safe withdrawal of immunosuppressive drugs has long been the goal of organ transplantation. We studied changes in the balance of T cells and myeloid cells in the blood of HLA-matched and -mismatched patients given living donor kidney transplants followed by total lymphoid irradiation, anti-thymocyte globulin conditioning, and donor hematopoietic cell transplant to induce mixed chimerism and immune tolerance. The clinical trials were based on a conditioning regimen used to establish mixed chimerism and tolerance in mice. In preclinical murine studies, there was a profound depletion of T cells and an increase in immunosuppressive polymorphonuclear (pmn) myeloid-derived suppressor cells (MDSCs) in the spleen and blood following transplant. Selective depletion of pmn MDSCs in mice abrogated mixed chimerism and tolerance. In our clinical trials, patients given an analogous tolerance conditioning regimen developed similar changes, including profound depletion of T cells and a marked increase in MDSCs in blood posttransplant. Posttransplant pmn MDSCs transiently increased expression of lectin-type oxidized LDL receptor-1, a marker of immunosuppression, and production of the T-cell inhibitor arginase-1. These posttransplant pmn MDSCs suppressed the activation, proliferation, and inflammatory cytokine secretion of autologous T-cell receptor microbead-stimulated pretransplant T cells when cocultured in vitro. In conclusion, we elucidated changes in receptors and function of immunosuppressive myeloid cells in patients enrolled in the tolerance protocol that were nearly identical to those of MDSCs required for tolerance in mice. These trials were registered at www.clinicaltrials.gov as #NCT00319657 and #NCT01165762.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Animaux , Essais cliniques comme sujet , Humains , Tolérance immunitaire , Souris , Cellules myéloïdes , Receveurs de transplantation , Conditionnement pour greffe
7.
PLoS One ; 15(11): e0242058, 2020.
Article de Anglais | MEDLINE | ID: mdl-33211709

RÉSUMÉ

The tumor immune microenvironment is emerging as a critical player in predicting cancer prognosis and response to therapies. However, the prognostic value of tumor-infiltrating immune cells in Gingivo-Buccal Oral Squamous Cell Carcinoma (GBOSCC) and their association with tumor size or lymph node metastases status require further elucidation. To study the relationship of tumor-infiltrating immune cells with tumor size (T stage) and lymph node metastases (N stages), we analyzed the density of tumor-infiltrating immune cells in archived, whole tumor resections from 94 patients. We characterized these sections by immune-histochemistry using 12 markers and enumerated tumor-infiltrating immune cells at the invasive margins (IM) and centers of tumors (CT). We observed that a higher density of CD3+ cells in the IM and CT was associated with smaller tumor size (T1-T2 stage). Fewer CD3+ cells was associated with larger tumor size (T3-T4 stage). High infiltration of CD3+and CD8+ cells in IM and CT as well as high CD4+ cell infiltrates in the IM was significantly associated with the absence of lymph node metastases. High infiltrates of CD3+ and CD8+ cells in CT was associated with significantly improved survival. Our results illustrate that the densities and spatial distribution of CD3+ and CD8+ cell infiltrates in primary GBOSCC tumors is predictive of disease progression and survival. Based on our findings, we recommend incorporating immune cell quantification in the TNM classification and routine histopathology reporting of GBOSCC. Immune cell quantification in CT and IM may help predict the efficacy of future therapies.


Sujet(s)
Antigènes CD3/métabolisme , Antigènes CD8/métabolisme , Carcinome épidermoïde/chirurgie , Lymphocytes TIL/immunologie , Tumeurs de la bouche/chirurgie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/anatomopathologie , Évolution de la maladie , Femelle , Humains , Lymphadénectomie , Métastase lymphatique , Mâle , Marges d'exérèse , Adulte d'âge moyen , Tumeurs de la bouche/immunologie , Tumeurs de la bouche/anatomopathologie , Stadification tumorale , Pronostic , Analyse de survie , Microenvironnement tumoral
8.
Radiat Res ; 194(6): 618-624, 2020 12 01.
Article de Anglais | MEDLINE | ID: mdl-32853385

RÉSUMÉ

Radiation therapy, along with surgery and chemotherapy, is one of the main treatments for cancer. While radiotherapy is highly effective in the treatment of localized tumors, its main limitation is its toxicity to normal tissue. Previous preclinical studies have reported that ultra-high dose-rate (FLASH) irradiation results in reduced toxicity to normal tissues while controlling tumor growth to a similar extent relative to conventional-dose-rate (CONV) irradiation. To our knowledge this is the first report of a dose-response study in mice comparing the effect of FLASH irradiation vs. CONV irradiation on skin toxicity. We found that FLASH irradiation results in both a lower incidence and lower severity of skin ulceration than CONV irradiation 8 weeks after single-fraction hemithoracic irradiation at high doses (30 and 40 Gy). Survival was also higher after FLASH hemithoracic irradiation (median survival >180 days at doses of 30 and 40 Gy) compared to CONV irradiation (median survival 100 and 52 days at 30 and 40 Gy, respectively). No ulceration was observed at doses 20 Gy or below in either FLASH or CONV. These results suggest a shifting of the dose-response curve for radiation-induced skin ulceration to the right for FLASH, compared to CONV irradiation, suggesting the potential for an enhanced therapeutic index for radiation therapy of cancer.


Sujet(s)
Radiothérapie/méthodes , Peau/effets des radiations , Animaux , Relation dose-effet des rayonnements , Femelle , Souris , Souris de lignée C57BL , Lésions radiques expérimentales/mortalité , Lésions radiques expérimentales/physiopathologie , Lésions radiques expérimentales/prévention et contrôle , Radiothérapie/effets indésirables , Indice de gravité de la maladie
9.
Semin Radiat Oncol ; 30(2): 194-200, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32381299

RÉSUMÉ

Radiation therapy benefits the majority of patients across the spectrum of cancer types. However, both local and distant tumor recurrences limit its clinical success. While departing from the established tenet of fractionation in clinical radiotherapy, ablative-intensity hypofractionated radiotherapy, especially stereotactic radiosurgery and stereotactic ablative radiotherapy, has emerged as an alternative paradigm achieving unprecedented rates of local tumor control. Direct tumor cell killing has been assumed to be the primary therapeutic mode of action of such ablative radiation. But with increasing recognition that tumor responses also depend on the immunostimulatory or immunosuppressive status of the tumor microenvironment, the immunologic effect of ablative radiotherapy is emerging as a key contributor to antitumor response. More recently, novel radiation modalities, such as spatially fractionated radiotherapy and ultrahigh dose rate FLASH irradiation, that venture even further from conventional paradigms have shown promise of increasing the therapeutic index of radiation therapy with the potential of immunomodulation. Here, we review the immunomodulatory impact of novel radiation therapy paradigms, heretofore considered radiobiological heresies, a deeper understanding of which is imperative to realizing fully their potential for more curative cancer therapy.


Sujet(s)
Immunomodulation/immunologie , Immunothérapie/tendances , Tumeurs/immunologie , Tumeurs/radiothérapie , Radio-oncologie/tendances , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des radiations , Association thérapeutique , Humains , Hypofractionnement de dose
10.
Curr Top Med Chem ; 18(22): 1976-1986, 2018.
Article de Anglais | MEDLINE | ID: mdl-30499391

RÉSUMÉ

Monensin is a lipid-soluble naturally occurring bioactive ionophore produced by Streptomyces spp. Its antimicrobial activity is mediated by its ability to exchange Na+ and K+ ions across the cell membrane thereby disrupting ionic gradients and altering cellular physiology. It is approved by Food and Drug Administration as a veterinary antibiotic to treat coccidiosis. Besides veterinary applications, monensin exhibits a broad spectrum activity against opportunistic pathogens of humans such as bacteria, virus, fungi and parasites in both drug sensitive and resistant strains. This ionophore can selectively kill pathogens with negligible toxic effect on mammalian cells. In this review, we discuss the therapeutic potential of monensin as a new broad-spectrum anti-microbial agent that warrants further studies for clinical use.


Sujet(s)
Antibactériens/pharmacologie , Antifongiques/pharmacologie , Antiparasitaires/pharmacologie , Monensin/pharmacologie , Animaux , Antibactériens/composition chimique , Antifongiques/composition chimique , Antiparasitaires/composition chimique , Bactéries/effets des médicaments et des substances chimiques , Champignons/effets des médicaments et des substances chimiques , Humains , Tests de sensibilité microbienne , Monensin/composition chimique , Parasites/effets des médicaments et des substances chimiques , Tests de sensibilité parasitaire
11.
Blood Adv ; 2(19): 2568-2580, 2018 10 09.
Article de Anglais | MEDLINE | ID: mdl-30301812

RÉSUMÉ

Conventional local tumor irradiation (LTI), delivered in small daily doses over several weeks, is used clinically as a palliative, rather than curative, treatment for chemotherapy-resistant diffuse large B-cell lymphoma (DLBCL) for patients who are ineligible for hematopoietic cell transplantation. Our goal was to test the hypothesis that accelerated, but not conventional, LTI would be more curative by inducing T cell-mediated durable remissions. We irradiated subcutaneous A20 and BL3750 lymphoma tumors in mice with a clinically relevant total radiation dose of 30 Gy LTI, delivered in 10 doses of 3 Gy over 4 days (accelerated irradiation) or as 10 doses of 3 Gy over 12 days (conventional irradiation). Compared with conventional LTI, accelerated LTI resulted in more complete and durable tumor remissions. The majority of these mice were resistant to rechallenge with lymphoma cells, demonstrating the induction of memory antitumor immunity. The increased efficacy of accelerated LTI correlated with higher levels of tumor cell necrosis vs apoptosis and expression of "immunogenic cell death" markers, including calreticulin, heat shock protein 70 (Hsp70), and Hsp90. Accelerated LTI-induced remissions were not seen in immunodeficient Rag-2 -/- mice, CD8+ T-cell-depleted mice, or Batf-3 -/- mice lacking CD8α+ and CD103+ dendritic cells. Accelerated, but not conventional, LTI in immunocompetent hosts induced marked increases in tumor-infiltrating CD4+ and CD8+ T cells and MHCII+CD103+CD11c+ dendritic cells and corresponding reductions in exhausted PD-1+Eomes+CD8+ T cells and CD4+CD25+FOXP3+ regulatory T cells. These findings raise the possibility that accelerated LTI can provide effective immune control of human DLBCL.


Sujet(s)
Lymphome B/immunologie , Lymphocytes T/immunologie , Animaux , Marqueurs biologiques , Cross-priming/immunologie , Cytokines/métabolisme , Cellules dendritiques/immunologie , Modèles animaux de maladie humaine , Humains , Immunité , Immunophénotypage , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Lymphocytes TIL/anatomopathologie , Lymphome B/mortalité , Lymphome B/anatomopathologie , Lymphome B/radiothérapie , Mâle , Souris , Souris knockout , Radiothérapie/méthodes , Induction de rémission , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Lymphocytes T/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Antimicrob Agents Chemother ; 60(3): 1304-18, 2015 Dec 14.
Article de Anglais | MEDLINE | ID: mdl-26666937

RÉSUMÉ

The global emergence of drug resistance in malaria is impeding the therapeutic efficacy of existing antimalarial drugs. Therefore, there is a critical need to develop an efficient drug delivery system to circumvent drug resistance. The anticoccidial drug monensin, a carboxylic ionophore, has been shown to have antimalarial properties. Here, we developed a liposome-based drug delivery of monensin and evaluated its antimalarial activity in lipid formulations of soya phosphatidylcholine (SPC) cholesterol (Chol) containing either stearylamine (SA) or phosphatidic acid (PA) and different densities of distearoyl phosphatidylethanolamine-methoxy-polyethylene glycol 2000 (DSPE-mPEG-2000). These formulations were found to be more effective than a comparable dose of free monensin in Plasmodium falciparum (3D7) cultures and established mice models of Plasmodium berghei strains NK65 and ANKA. Parasite killing was determined by a radiolabeled [(3)H]hypoxanthine incorporation assay (in vitro) and microscopic counting of Giemsa-stained infected erythrocytes (in vivo). The enhancement of antimalarial activity was dependent on the liposomal lipid composition and preferential uptake by infected red blood cells (RBCs). The antiplasmodial activity of monensin in SA liposome (50% inhibitory concentration [IC50], 0.74 nM) and SPC:Chol-liposome with 5 mol% DSPE-mPEG 2000 (IC50, 0.39 nM) was superior to that of free monensin (IC50, 3.17 nM), without causing hemolysis of erythrocytes. Liposomes exhibited a spherical shape, with sizes ranging from 90 to 120 nm, as measured by dynamic light scattering and high-resolution electron microscopy. Monensin in long-circulating liposomes of stearylamine with 5 mol% DSPE-mPEG 2000 in combination with free artemisinin resulted in enhanced killing of parasites, prevented parasite recrudescence, and improved survival. This is the first report to demonstrate that monensin in PEGylated stearylamine (SA) liposome has therapeutic potential against malaria infections.


Sujet(s)
Antipaludiques/pharmacologie , Artémisinines/pharmacologie , Liposomes/administration et posologie , Paludisme/traitement médicamenteux , Monensin/pharmacologie , Amines/administration et posologie , Amines/composition chimique , Animaux , Antipaludiques/administration et posologie , Sang/effets des médicaments et des substances chimiques , Sang/parasitologie , Systèmes de délivrance de médicaments/méthodes , Association de médicaments , Femelle , Liposomes/composition chimique , Liposomes/pharmacologie , Paludisme/parasitologie , Souris , Monensin/pharmacocinétique , Plasmodium berghei/pathogénicité , Plasmodium falciparum/effets des médicaments et des substances chimiques , Distribution tissulaire
13.
Clin Immunol ; 161(2): 355-65, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26499378

RÉSUMÉ

Schimke immuno-osseous dysplasia (SIOD) is an autosomal recessive, fatal childhood disorder associated with skeletal dysplasia, renal dysfunction, and T-cell immunodeficiency. This disease is linked to biallelic loss-of-function mutations of the SMARCAL1 gene. Although recurrent infection, due to T-cell deficiency, is a leading cause of morbidity and mortality, the etiology of the T-cell immunodeficiency is unclear. Here, we demonstrate that the T cells of SIOD patients have undetectable levels of protein and mRNA for the IL-7 receptor alpha chain (IL7Rα) and are unresponsive to stimulation with IL-7, indicating a loss of functional receptor. No pathogenic mutations were detected in the exons of IL7R in these patients; however, CpG sites in the IL7R promoter were hypermethylated in SIOD T cells. We propose therefore that the lack of IL7Rα expression, associated with hypermethylation of the IL7R promoter, in T cells and possibly their earlier progenitors, restricts T-cell development in SIOD patients.


Sujet(s)
Artériosclérose/génétique , Déficits immunitaires/génétique , Syndrome néphrotique/génétique , Ostéochondrodysplasies/génétique , Embolie pulmonaire/génétique , Récepteurs à l'interleukine-7/génétique , Lymphocytes T/métabolisme , Adolescent , Adulte , Artériosclérose/métabolisme , Artériosclérose/anatomopathologie , Cellules cultivées , Enfant , Enfant d'âge préscolaire , Helicase/génétique , Méthylation de l'ADN , Cytométrie en flux , Expression des gènes , Humains , Immunohistochimie , Déficits immunitaires/métabolisme , Déficits immunitaires/anatomopathologie , Interleukine-17/pharmacologie , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Mutation , Syndrome néphrotique/métabolisme , Syndrome néphrotique/anatomopathologie , Ostéochondrodysplasies/métabolisme , Ostéochondrodysplasies/anatomopathologie , Maladies d'immunodéficience primaire , Régions promotrices (génétique)/génétique , Embolie pulmonaire/métabolisme , Embolie pulmonaire/anatomopathologie , Récepteurs à l'interleukine-7/métabolisme , RT-PCR , Analyse de séquence d'ADN , Jeune adulte
14.
Clin Cancer Res ; 21(16): 3727-39, 2015 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-25869387

RÉSUMÉ

PURPOSE: The goals of the study were to elucidate the immune mechanisms that contribute to desirable complete remissions of murine colon tumors treated with single radiation dose of 30 Gy. This dose is at the upper end of the ablative range used clinically to treat advanced or metastatic colorectal, liver, and non-small cell lung tumors. EXPERIMENTAL DESIGN: Changes in the tumor immune microenvironment of single tumor nodules exposed to radiation were studied using 21-day (>1 cm in diameter) CT26 and MC38 colon tumors. These are well-characterized weakly immunogenic tumors. RESULTS: We found that the high-dose radiation transformed the immunosuppressive tumor microenvironment resulting in an intense CD8(+) T-cell tumor infiltrate, and a loss of myeloid-derived suppressor cells (MDSC). The change was dependent on antigen cross-presenting CD8(+) dendritic cells, secretion of IFNγ, and CD4(+)T cells expressing CD40L. Antitumor CD8(+) T cells entered tumors shortly after radiotherapy, reversed MDSC infiltration, and mediated durable remissions in an IFNγ-dependent manner. Interestingly, extended fractionated radiation regimen did not result in robust CD8(+) T-cell infiltration. CONCLUSIONS: For immunologically sensitive tumors, these results indicate that remissions induced by a short course of high-dose radiotherapy depend on the development of antitumor immunity that is reflected by the nature and kinetics of changes induced in the tumor cell microenvironment. These results suggest that systematic examination of the tumor immune microenvironment may help in optimizing the radiation regimen used to treat tumors by adding a robust immune response.


Sujet(s)
Tumeurs du côlon/immunologie , Tumeurs du côlon/radiothérapie , Microenvironnement tumoral/immunologie , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/effets des radiations , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des radiations , Lignée cellulaire tumorale , Tumeurs du côlon/anatomopathologie , Cellules dendritiques/immunologie , Cellules dendritiques/effets des radiations , Humains , Souris , Cellules myéloïdes/immunologie , Cellules myéloïdes/effets des radiations , Induction de rémission , Microenvironnement tumoral/effets des radiations
15.
Clin Cancer Res ; 21(14): 3113-20, 2015 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-25908780

RÉSUMÉ

In the past 5 years, immunomodulatory antibodies have revolutionized cancer immunotherapy. CD137, a member of the tumor necrosis factor receptor superfamily, represents a promising target for enhancing antitumor immune responses. CD137 helps regulate the activation of many immune cells, including CD4(+) T cells, CD8(+) T cells, dendritic cells, and natural killer cells. Recent studies indicate that the antitumor efficacy of therapeutic tumor-targeting antibodies can be augmented by the addition of agonistic antibodies targeting CD137. As ligation of CD137 provides a costimulatory signal in multiple immune cell subsets, combination therapy of CD137 antibody with therapeutic antibodies and/or vaccination has the potential to improve cancer treatment. Recently, clinical trials of combination therapies with agonistic anti-CD137 mAbs have been launched. In this review, we discuss the recent advances and clinical promise of agonistic anti-CD137 monoclonal antibody therapy.


Sujet(s)
Anticorps monoclonaux/immunologie , Immunothérapie/méthodes , Tumeurs/thérapie , Antigènes CD137/immunologie , Animaux , Anticorps monoclonaux/pharmacologie , Humains
16.
Radiat Res ; 182(2): 163-9, 2014 Aug.
Article de Anglais | MEDLINE | ID: mdl-24992165

RÉSUMÉ

The goal of this study was to determine whether a combination of local tumor irradiation and autologous T-cell transplantation can effectively treat metastatic 4T1 breast cancer in mice. BALB/c mice were injected subcutaneously with luciferase-labeled 4T1 breast tumor cells and allowed to grow for 21 days, at which time metastases appeared in the lungs. Primary tumors were treated at that time with 3 daily fractions of 20 Gy of radiation each. Although this approach could eradicate primary tumors, tumors in the lungs grew progressively. We attempted to improve efficacy of the radiation by adding autologous T-cell infusions. Accordingly, T cells were purified from the spleens of tumor-bearing mice after completion of irradiation and cryopreserved. Cyclophosphamide was administered thereafter to induce lymphodepletion, followed by T-cell infusion. Although the addition of cyclophosphamide to irradiation did not improve survival or reduce tumor progression, the combination of radiation, cyclophosphamide and autologous T-cell infusion induced durable remissions and markedly improved survival. We conclude that the combination of radiation and autologous T-cell infusion is an effective treatment for metastatic 4T1 breast cancer.


Sujet(s)
Fractionnement de la dose d'irradiation , Tumeurs expérimentales de la mamelle/immunologie , Tumeurs expérimentales de la mamelle/radiothérapie , Lymphocytes T/transplantation , Animaux , Lignée cellulaire tumorale , Association thérapeutique , Cyclophosphamide/pharmacologie , Femelle , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris de lignée BALB C , Métastase tumorale , Sous-populations de lymphocytes T/effets des médicaments et des substances chimiques , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/transplantation , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Transplantation autologue
17.
Blood ; 119(6): 1581-9, 2012 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-22174155

RÉSUMÉ

We used a model of combined bone marrow and heart transplantation, in which tolerance and stable chimerism is induced after conditioning with fractionated irradiation of the lymphoid tissues and anti-T-cell antibodies. Graft acceptance and chimerism required host CD4(+)CD25(+) Treg production of IL-10 that was in-turn enhanced by host invariant natural killer (NK) T-cell production of IL-4. Up-regulation of PD-1 on host Tregs, CD4(+)CD25(-) conventional T (Tcon) cells, and CD8(+) T cells was also enhanced by NKT cell production of IL-4. Up-regulated PD-1 expression on Tregs was linked to IL-10 secretion, on CD8(+) T cells was linked to Tim-3 expression, and on CD4(+) Tcon cells was associated with reduced IFNγ secretion. Changes in the expression of PD-1 were induced by the conditioning regimen, and declined after bone marrow transplantation. In conclusion, NKT cells in this model promoted changes in expression of negative costimulatory receptors and anti-inflammatory cytokines by Tregs and other T-cell subsets in an IL-4-dependent manner that resulted in tolerance to the bone marrow and organ grafts.


Sujet(s)
Transplantation de moelle osseuse/immunologie , Transplantation cardiaque/immunologie , Tolérance immunitaire/immunologie , Cellules T tueuses naturelles/immunologie , Lymphocytes T régulateurs/immunologie , Animaux , Antigènes de différenciation/immunologie , Antigènes de différenciation/métabolisme , Sérum antilymphocyte/pharmacologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Communication cellulaire/immunologie , Cellules cultivées , Femelle , Technique d'immunofluorescence , Récepteur cellulaire-2 du virus de l'hépatite A , Interféron gamma/immunologie , Interféron gamma/métabolisme , Interleukine-10/génétique , Interleukine-10/immunologie , Interleukine-10/métabolisme , Interleukine-4/génétique , Interleukine-4/immunologie , Interleukine-4/métabolisme , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout , Cellules T tueuses naturelles/métabolisme , Récepteur-1 de mort cellulaire programmée , Récepteurs viraux/immunologie , Récepteurs viraux/métabolisme , Lymphocytes T régulateurs/métabolisme , Conditionnement pour greffe/méthodes , Régulation positive/effets des médicaments et des substances chimiques
18.
Blood ; 118(19): 5319-29, 2011 Nov 10.
Article de Anglais | MEDLINE | ID: mdl-21868578

RÉSUMÉ

The curative potential of MHC-matched allogeneic bone marrow transplantation (BMT) is in part because of immunologic graft-versus-tumor (GvT) reactions mediated by donor T cells that recognize host minor histocompatibility antigens. Immunization with leukemia-associated antigens, such as Wilms Tumor 1 (WT1) peptides, induces a T-cell population that is tumor antigen specific. We determined whether allogeneic BMT combined with immunotherapy using WT1 peptide vaccination of donors induced more potent antitumor activity than either therapy alone. WT1 peptide vaccinations of healthy donor mice induced CD8(+) T cells that were specifically reactive to WT1-expressing FBL3 leukemia cells. We found that peptide immunization was effective as a prophylactic vaccination before tumor challenge, yet was ineffective as a therapeutic vaccination in tumor-bearing mice. BMT from vaccinated healthy MHC-matched donors, but not syngeneic donors, into recipient tumor-bearing mice was effective as a therapeutic maneuver and resulted in eradication of FBL3 leukemia. The transfer of total CD8(+) T cells from immunized donors was more effective than the transfer of WT1-tetramer(+)CD8(+) T cells and both required CD4(+) T-cell help for maximal antitumor activity. These findings show that WT1 peptide vaccination of donor mice can dramatically enhance GvT activity after MHC-matched allogeneic BMT.


Sujet(s)
Transplantation de moelle osseuse/immunologie , Réaction du greffon contre la leucémie/immunologie , Leucémie expérimentale/immunologie , Leucémie expérimentale/thérapie , Protéines WT1/immunologie , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/transplantation , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/transplantation , Immunisation , Mémoire immunologique , Immunothérapie adoptive , Complexe majeur d'histocompatibilité , Mâle , Souris , Souris de lignée C3H , Souris de lignée C57BL , Donneurs de tissus , Transplantation homologue , Transplantation isogénique , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/immunologie
19.
Blood ; 117(11): 3230-9, 2011 Mar 17.
Article de Anglais | MEDLINE | ID: mdl-21239702

RÉSUMÉ

Allogeneic hematopoietic cell transplantation can be curative in patients with leukemia and lymphoma. However, progressive growth of malignant cells, relapse after transplantation, and graft-versus-host disease (GVHD) remain important problems. The goal of the current murine study was to select a freshly isolated donor T-cell subset for infusion that separates antilymphoma activity from GVHD, and to determine whether the selected subset could effectively prevent or treat progressive growth of a naturally occurring B-cell lymphoma (BCL(1)) without GVHD after recipients were given T cell-depleted bone marrow transplantations from major histocompatibility complex-mismatched donors. Lethal GVHD was observed when total T cells, naive CD4(+) T cells, or naive CD8(+) T cells were used. Memory CD4(+)CD44(hi) and CD8(+)CD44(hi) T cells containing both central and effector memory cells did not induce lethal GVHD, but only memory CD8(+) T cells had potent antilymphoma activity and promoted complete chimerism. Infusion of CD8(+) memory T cells after transplantation was able to eradicate the BCL(1) lymphoma even after progressive growth without inducing severe GVHD. In conclusion, the memory CD8(+) T-cell subset separated graft antilymphoma activity from GVHD more effectively than naive T cells, memory CD4(+) T cells, or memory total T cells.


Sujet(s)
Transplantation de moelle osseuse , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Maladie du greffon contre l'hôte/immunologie , Antigènes CD44/métabolisme , Mémoire immunologique/immunologie , Lymphomes/immunologie , Animaux , Poids , Lymphocytes T CD4+/cytologie , Lymphocytes T CD8+/cytologie , Division cellulaire , Lignée cellulaire tumorale , Évolution de la maladie , Maladie du greffon contre l'hôte/complications , Immunisation , Lymphomes/complications , Lymphomes/thérapie , Souris , Spécificité d'organe/immunologie , Phénotype , Rate/cytologie , Rate/immunologie , Analyse de survie , Sous-populations de lymphocytes T/transplantation
20.
Blood ; 113(18): 4458-67, 2009 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-19221040

RÉSUMÉ

Although CD4(+)CD25(+) T cells (T regulatory cells [Tregs]) and natural killer T cells (NKT cells) each protect against graft-versus-host disease (GVHD), interactions between these 2 regulatory cell populations after allogeneic bone marrow transplantation (BMT) have not been studied. We show that host NKT cells can induce an in vivo expansion of donor Tregs that prevents lethal GVHD in mice after conditioning with fractionated lymphoid irradiation (TLI) and anti-T-cell antibodies, a regimen that models human GVHD-protective nonmyeloablative protocols using TLI and antithymocyte globulin (ATG), followed by allogeneic hematopoietic cell transplantation (HCT). GVHD protection was lost in NKT-cell-deficient Jalpha18(-/-) hosts and interleukin-4 (IL-4)(-/-) hosts, or when the donor transplant was Treg depleted. Add-back of donor Tregs or wild-type host NKT cells restored GVHD protection. Donor Treg proliferation was lost in IL-4(-/-) hosts or when IL-4(-/-) mice were used as the source of NKT cells for adoptive transfer, indicating that host NKT cell augmentation of donor Treg proliferation after TLI/antithymocyte serum is IL-4 dependent. Our results demonstrate that host NKT cells and donor Tregs can act synergistically after BMT, and provide a mechanism by which strategies designed to preserve host regulatory cells can augment in vivo donor Treg expansion to regulate GVHD after allogeneic HCT.


Sujet(s)
Maladie du greffon contre l'hôte/immunologie , Maladie du greffon contre l'hôte/prévention et contrôle , Interleukine-4/immunologie , Cellules T tueuses naturelles/immunologie , Lymphocytes T régulateurs/immunologie , Transfert adoptif , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Prolifération cellulaire , Cytokines/métabolisme , Cytométrie en flux , Facteurs de transcription Forkhead/métabolisme , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Rate/immunologie , Rate/métabolisme , Lymphocytes T régulateurs/cytologie , Lymphocytes T régulateurs/transplantation , Donneurs de tissus , Irradiation corporelle totale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE