Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
1.
Ultrasound Med Biol ; 49(9): 2095-2102, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37365065

RÉSUMÉ

OBJECTIVE: B-lines are a ring-down artifact of lung ultrasound that arise with increased alveolar water in conditions such as pulmonary edema and infectious pneumonitis. Confluent B-line presence may signify a different level of pathology compared with single B-lines. Existing algorithms aimed at B-line counting do not distinguish between single and confluent B-lines. The objective of this study was to test a machine learning algorithm for confluent B-line identification. METHODS: This study used a subset of 416 clips from 157 subjects, previously acquired in a prospective study enrolling adults with shortness of breath at two academic medical centers, using a hand-held tablet and a 14-zone protocol. After exclusions, random sampling generated a total of 416 clips (146 curvilinear, 150 sector and 120 linear) for review. A group of five experts in point-of-care ultrasound blindly evaluated the clips for presence/absence of confluent B-lines. Ground truth was defined as majority agreement among the experts and used for comparison with the algorithm. RESULTS: Confluent B-lines were present in 206 of 416 clips (49.5%). Sensitivity and specificity of confluent B-line detection by algorithm compared with expert determination were 83% (95% confidence interval [CI]: 0.77-0.88) and 92% (95% CI: 0.88-0.96). Sensitivity and specificity did not statistically differ between transducers. Agreement between algorithm and expert for confluent B-lines measured by unweighted κ was 0.75 (95% CI: 0.69-0.81) for the overall set. CONCLUSION: The confluent B-line detection algorithm had high sensitivity and specificity for detection of confluent B-lines in lung ultrasound point-of-care clips, compared with expert determination.


Sujet(s)
Poumon , Oedème pulmonaire , Adulte , Humains , Études prospectives , Poumon/imagerie diagnostique , Algorithmes , Échographie/méthodes , Apprentissage machine
2.
Expert Rev Mol Diagn ; 21(3): 333-342, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33849372

RÉSUMÉ

Background: Here we compare the performance of the high-throughput BD COR System (COR) to the Viper LT System (Viper) using the BD Onclarity HPV assay.Research Design and Methods: Remnant clinical specimens, contrived specimens in SurePath (BD) and PreservCyt (Hologic) media, and prospective clinical specimens in BD Cervical Brush Diluent (CBD) were tested. Outcomes included intra-laboratory agreement of Onclarity results on COR and inter-system agreement between COR and Viper.Results: Onclarity reproducibility on COR resulted in standard deviation and correlation of variation of Ct values ranging from 0.14 to 1.98 and 0.49% to 2.15%, respectively, for contrived specimens, and 0.9-3.08 and 2.89-9.21%, respectively, for clinical specimens. In the COR and Viper clinical agreement study, OPA for Onclarity ranged from 97.1%-98.9%, depending on the collection media type. PPA values for pooled, HPV(+) specimens at low positive (C95), and moderate positive (3XC95) target concentrations were ≥95.0% and 100%, respectively; PPA values associated with HPV 16, 18, 31, 45, 33/58, 52, 35/39/68, 51, and 56/59/66, individually, ranged from 93.8%-100%.Conclusions: Onclarity performance on COR is equivalent to Viper, and is accurate and reproducible for detection of all high-risk HPV genotypes, with a throughput of 330 results from a single 8-hour shift.


Sujet(s)
Infections à papillomavirus , Tumeurs du col de l'utérus , Femelle , Humains , Papillomaviridae/génétique , Infections à papillomavirus/diagnostic , Études prospectives , Reproductibilité des résultats , Sensibilité et spécificité , Tumeurs du col de l'utérus/diagnostic
3.
Gastroenterology ; 157(3): 838-850.e6, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31163177

RÉSUMÉ

BACKGROUND & AIMS: Little is known about mechanisms of perineural invasion (PNI) by pancreatic ductal adenocarcinomas (PDAs) or other tumors. Annexin A2 (ANXA2) regulates secretion of SEMA3D, an axon guidance molecule, which binds and activates the receptor PLXND1 to promote PDA invasion and metastasis. We investigated whether axon guidance molecules promote PNI and metastasis by PDA cells in mice. METHODS: We performed studies in a dorsal root ganglion (DRG) invasion system, wild-type C57BL/6 mice (controls), mice with peripheral sensory neuron-specific disruption of PlxnD1 (PLAC mice), LSL-KRASG12D/+;LSL-TP53R172H/+;PDX-1-CRE+/+ (KPC) mice, and KPC mice crossed with ANXA2-knockout mice (KPCA mice). PDA cells were isolated from KPC mice and DRG cells were isolated from control mice. Levels of SEMA3D or ANXA2 were knocked down in PDA cells with small hairpin and interfering RNAs and cells were analyzed by immunoblots in migration assays, with DRGs and with or without antibodies against PLXND1. PDA cells were injected into the pancreas of control and PLAC mice, growth of tumors was assessed, and tumor samples were analyzed by histology. DRG cells were incubated with SEMA3D and analyzed by live imaging. We measured levels of SEMA3D and PLXND1 in PDA specimens from patients with PNI and calculated distances between tumor cells and nerves. RESULTS: DRG cells increase the migration of PDC cells in invasion assays; knockdown of SEMA3D in PDA cells or antibody blockade of PLXND1 on DRG cells reduced this invasive activity. In mice, orthotopic tumors grown from PDA cells with knockdown of SEMA3D, and in PLAC mice, orthotopic tumors grown from PDA cells, had reduced innervation and formed fewer metastases than orthotopic tumors grown from PDA cells in control mice. Increased levels of SEMA3D and PLXND1 in human PDA specimens associated with PNI. CONCLUSIONS: DRG cells increase the migratory and invasive activities of pancreatic cancer cells, via secretion of SEMA3D by pancreatic cells and activation of PLXND1 on DRGs. Knockdown of SEMA3D and loss of neural PLXND1 reduces innervation of orthotopic PDAs and metastasis in mice. Increased levels of SEMA3D and PLXND1 in human PDA specimens associated with PNI. Strategies to disrupt the axon guidance pathway mediated by SEMA3D and PLXND1 might be developed to slow progression of PDA.


Sujet(s)
Annexine A2/métabolisme , Guidage axonal , Carcinome du canal pancréatique/métabolisme , Mouvement cellulaire , Ganglions sensitifs des nerfs spinaux/métabolisme , Glycoprotéines membranaires/métabolisme , Protéines de tissu nerveux/métabolisme , Tumeurs du pancréas/métabolisme , Sémaphorines/métabolisme , Animaux , Annexine A2/déficit , Annexine A2/génétique , Guidage axonal/génétique , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/secondaire , Communication cellulaire , Ganglions sensitifs des nerfs spinaux/anatomopathologie , Régulation de l'expression des gènes tumoraux , Gènes p53 , Gènes ras , Prédisposition génétique à une maladie , Protéines à homéodomaine/génétique , Humains , Protéines et peptides de signalisation intracellulaire , Glycoprotéines membranaires/déficit , Glycoprotéines membranaires/génétique , Souris de souche-129 , Souris de lignée C57BL , Souris knockout , Invasion tumorale , Protéines de tissu nerveux/déficit , Protéines de tissu nerveux/génétique , Excroissance neuronale , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Phénotype , Sémaphorines/génétique , Transduction du signal , Transactivateurs/génétique , Cellules cancéreuses en culture
4.
Mol Cancer Ther ; 16(11): 2399-2409, 2017 11.
Article de Anglais | MEDLINE | ID: mdl-28864680

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is one of the most chemotherapy- and radiotherapy-resistant tumors. The c-Met and Hedgehog (Hh) pathways have been shown previously by our group to be key regulatory pathways in the primary tumor growth and metastases formation. Targeting both the HGF/c-Met and Hh pathways has shown promising results in preclinical studies; however, the benefits were not readily translated into clinical trials with PDAC patients. In this study, utilizing mouse models of PDAC, we showed that inhibition of either HGF/c-Met or Hh pathways sensitize the PDAC tumors to gemcitabine, resulting in decreased primary tumor volume as well as significant reduction of metastatic tumor burden. However, prolonged treatment of single HGF/c-Met or Hh inhibitor leads to resistance to these single inhibitors, likely because the single c-Met treatment leads to enhanced expression of Shh, and vice versa. Targeting both the HGF/c-Met and Hh pathways simultaneously overcame the resistance to the single-inhibitor treatment and led to a more potent antitumor effect in combination with the chemotherapy treatment. Mol Cancer Ther; 16(11); 2399-409. ©2017 AACR.


Sujet(s)
Adénocarcinome/traitement médicamenteux , Carcinome du canal pancréatique/traitement médicamenteux , Facteur de croissance des hépatocytes/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Animaux , Benzamides/administration et posologie , Dérivés du biphényle/administration et posologie , Composés hétérocycliques bicycliques/administration et posologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Désoxycytidine/administration et posologie , Désoxycytidine/analogues et dérivés , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines Hedgehog/antagonistes et inhibiteurs , Protéines Hedgehog/génétique , Facteur de croissance des hépatocytes/génétique , Humains , Imidazoles , Souris , Souris transgéniques , Métastase tumorale , Protéines proto-oncogènes c-met/génétique , Pyridines/administration et posologie , Triazines , Microenvironnement tumoral/effets des médicaments et des substances chimiques ,
5.
J Gastrointest Oncol ; 8(3): 441-448, 2017 Jun.
Article de Anglais | MEDLINE | ID: mdl-28736631

RÉSUMÉ

Pancreatic adenocarcinoma is still widely considered as a deadly disease even though there are substantial therapeutic developments in the past decade. Using combinational chemotherapy regimens, represented by gemcitabine plus nab-paclitaxel and FOLFIRINOX, was able to improve overall survival in patients with advanced disease to a limited extent. It has been a challenge to develop targeted therapies that are focused on the neoplasm cells of pancreatic adenocarcinoma. Recently, targeting the stroma and immune compartments of pancreatic adenocarcinoma has shown promising results. The paradigm of biologics drug development therefore has been shifted by extending to these exciting areas. Although some of the preclinical and clinical researches in targeting the tumor microenvironment of pancreatic adenocarcinoma have shown promising results, others have resulted in controversial findings. Both comprehensive and in-depth researches on the basic science of the tumor microenvironment of pancreatic adenocarcinoma are thus warranted for the development of effective biologics that target the tumor microenvironment. Moreover, an ideal treatment for pancreatic adenocarcinoma shall be a combination of targeting both neoplastic cells and the tumor microenvironment.

6.
Science ; 357(6349): 409-413, 2017 07 28.
Article de Anglais | MEDLINE | ID: mdl-28596308

RÉSUMÉ

The genomes of cancers deficient in mismatch repair contain exceptionally high numbers of somatic mutations. In a proof-of-concept study, we previously showed that colorectal cancers with mismatch repair deficiency were sensitive to immune checkpoint blockade with antibodies to programmed death receptor-1 (PD-1). We have now expanded this study to evaluate the efficacy of PD-1 blockade in patients with advanced mismatch repair-deficient cancers across 12 different tumor types. Objective radiographic responses were observed in 53% of patients, and complete responses were achieved in 21% of patients. Responses were durable, with median progression-free survival and overall survival still not reached. Functional analysis in a responding patient demonstrated rapid in vivo expansion of neoantigen-specific T cell clones that were reactive to mutant neopeptides found in the tumor. These data support the hypothesis that the large proportion of mutant neoantigens in mismatch repair-deficient cancers make them sensitive to immune checkpoint blockade, regardless of the cancers' tissue of origin.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Marqueurs biologiques tumoraux/antagonistes et inhibiteurs , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Tumeurs colorectales/immunologie , Tumeurs colorectales/thérapie , Syndromes néoplasiques héréditaires/immunologie , Syndromes néoplasiques héréditaires/thérapie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes néoplasiques/immunologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/mortalité , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Tumeurs colorectales/génétique , Tumeurs colorectales/mortalité , Réparation de mésappariement de l'ADN , Survie sans rechute , Femelle , Humains , Mâle , Adulte d'âge moyen , Mutation , Syndromes néoplasiques héréditaires/génétique , Syndromes néoplasiques héréditaires/mortalité , Récepteur-1 de mort cellulaire programmée/immunologie , Lymphocytes T/immunologie , Jeune adulte
7.
Oncotarget ; 8(63): 106405-106414, 2017 Dec 05.
Article de Anglais | MEDLINE | ID: mdl-29290958

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDA) is renowned for high rates of metastasis and poor survival. Its notoriously dense fibrotic stroma contributes to chemoresistance. Stromal signaling in PDA is recognized for its multiple roles in regulating tumor invasion and metastasis. However, no stromal biomarker which can predict survival in PDA exists. Annexin A2 (AnxA2) was formerly identified as a metastasis-associated protein in PDA and tumoral overexpression is associated with poor survival. In this study, we examined AnxA2 expression in the tumor microenvironment in a preclinical model of PDA which suggests its role in tumor colonization. We injected wild-type (KPC) and AnxA2 knockout (KPCA) pancreatic cells into C57BL/GJ (B6) and AnxA2 knockout (KO) mice using the hemi-spleen model and observed their survival. We performed quantitative immunohistochemistry examining stromal AnxA2 expression in 56 patients who had surgically resected PDA and correlated expression with clinical outcomes. B6 mice injected with KPC cells demonstrated decreased median survival compared to those injected with KPCA cells (90 days vs. not reached, p < 0.0001) whereas there was no survival difference in the AnxA2 KO mice (p = 0.63). In patient specimens, we found that high stromal AnxA2 expression (≥80th percentile) was associated with significantly reduced disease-free survival (p = 0.002) and overall survival (p < 0.001). Using multivariate Cox models, there were no significant associations between other clinical covariates apart from high stromal AnxA2 expression. This study highlights the role that stromal AnxA2 expression plays as a prognostic marker in PDA and its potential as a predictive biomarker for survival outcomes in PDA.

8.
Cancer Res ; 77(1): 41-52, 2017 01 01.
Article de Anglais | MEDLINE | ID: mdl-27821486

RÉSUMÉ

Understanding how stromal signals regulate the development of pancreatic ductal adenocarcinoma (PDAC) may suggest novel therapeutic interventions in this disease. In this study, we assessed the metastatic role of stromal signals suggested to be important in the PDAC microenvironment. Src and IGF-1R phosphorylated the prometastatic molecule Annexin A2 (AnxA2) at Y23 and Y333 in response to stromal signals HGF and IGF-1, respectively, and IGF-1 expression was regulated by the Sonic Hedgehog (Shh) pathway. Both Shh and HGF were heterogeneously expressed in PDAC stroma, and only dual inhibition of these pathways could significantly suppress AnxA2 phosphorylation, PDAC growth, and metastasis. Taken together, our results illuminate tumor-stromal interactions, which drive metastasis, and provide a mechanism-based rationale for a stroma-directed therapy for PDAC. Cancer Res; 77(1); 41-52. ©2016 AACR.


Sujet(s)
Carcinome du canal pancréatique/anatomopathologie , Régulation de l'expression des gènes tumoraux/physiologie , Invasion tumorale/anatomopathologie , Tumeurs du pancréas/anatomopathologie , Transduction du signal/physiologie , Microenvironnement tumoral/physiologie , Animaux , Technique de Western , Chromatographie en phase liquide , Test ELISA , Hétérogreffes , Humains , Immunohistochimie , Souris , Réaction de polymérisation en chaine en temps réel , Cellules stromales/métabolisme , Spectrométrie de masse en tandem
9.
Cancer Res ; 76(18): 5395-404, 2016 09 15.
Article de Anglais | MEDLINE | ID: mdl-27496707

RÉSUMÉ

Stromal fibrosis is a prominent histologic characteristic of pancreatic ductal adenocarcinoma (PDAC), but how stromal fibroblasts are regulated in the tumor microenvironment (TME) to support tumor growth is largely unknown. Here we show that PDAC cells can induce DNA methylation in cancer-associated fibroblasts (CAF). Upon direct contact with PDAC cells, DNA methylation of SOCS1 and other genes is induced in mesenchymal stem cells or in CAF that lack SOCS1 methylation at baseline. Silencing or decitabine treatment to block the DNA methylation enzyme DNMT1 inhibited methylation of SOCS1. In contrast, SOCS1 gene methylation and downregulation in CAF activated STAT3 and induced insulin-like growth factor-1 expression to support PDAC cell growth. Moreover, CAF facilitated methylation-dependent growth of PDAC tumor xenografts in mice. The ability of patient-derived CAF with SOCS1 methylation to promote PDAC growth was more robust than CAF without SOCS1 methylation. Overall, our results reveal how PDAC cells can reprogram CAF to modify tumor-stromal interactions in the TME, which promote malignant growth and progression. Cancer Res; 76(18); 5395-404. ©2016 AACR.


Sujet(s)
Fibroblastes associés au cancer/anatomopathologie , Carcinome du canal pancréatique/génétique , Méthylation de l'ADN/génétique , Tumeurs du pancréas/génétique , Protéine-1 suppressive de la signalisation des cytokines/génétique , Animaux , Carcinome du canal pancréatique/anatomopathologie , Hétérogreffes , Humains , Souris , Souris de lignée NOD , Souris SCID , Séquençage par oligonucléotides en batterie , Tumeurs du pancréas/anatomopathologie , Réaction de polymérisation en chaîne , Microenvironnement tumoral
10.
Oncotarget ; 6(40): 43005-15, 2015 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-26515728

RÉSUMÉ

Our neoadjuvant clinical trial of a GM-CSF secreting allogeneic pancreas tumor vaccine (GVAX) revealed the development of tertiary lymphoid aggregates (TLAs) within the pancreatic ductal adenocarcinoma (PDA) tumor microenvironment 2 weeks after GVAX treatment. Microarray studies revealed that multiple components of the TGF-ß pathway were suppressed in TLAs from patients who survived greater than 3 years and who demonstrated vaccine-enhanced mesothelin-specific T cell responses. We tested the hypothesis that combining GVAX with TGF-ß inhibitors will improve the anti-tumor immune response of vaccine therapy. In a metastatic murine model of pancreatic cancer, combination therapy with GVAX vaccine and a TGF-ß blocking antibody improved the cure rate of PDA-bearing mice. TGF-ß blockade in combination with GVAX significantly increased the infiltration of effector CD8+ T lymphocytes, specifically anti-tumor-specific IFN-g producing CD8+ T cells, when compared to monotherapy controls (all p < 0.05). TGF-ß blockade alone did not deplete T regulatory cells (Tregs), but when give in combination with GVAX, GVAX induced intratumoral Tregs were depleted. Therefore, our PDA preclinical model demonstrates a survival advantage in mice treated with an anti-TGF-ß antibody combined with GVAX therapy and provides strong rational for testing this combinational therapy in clinical trials.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Vaccins anticancéreux/administration et posologie , Carcinome du canal pancréatique/immunologie , Tumeurs du pancréas/immunologie , Lymphocytes T régulateurs/immunologie , Animaux , Anticorps neutralisants/administration et posologie , Carcinome du canal pancréatique/anatomopathologie , Modèles animaux de maladie humaine , Test ELISA , Cytométrie en flux , Lymphocytes TIL/effets des médicaments et des substances chimiques , Mésothéline , Souris , Tumeurs du pancréas/anatomopathologie , Facteur de croissance transformant bêta/antagonistes et inhibiteurs
11.
Sci Signal ; 8(388): ra77, 2015 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-26243191

RÉSUMÉ

Most patients with pancreatic ductal adenocarcinoma (PDA) present with metastatic disease at the time of diagnosis or will recur with metastases after surgical treatment. Semaphorin-plexin signaling mediates the migration of neuronal axons during development and of blood vessels during angiogenesis. The expression of the gene encoding semaphorin 3D (Sema3D) is increased in PDA tumors, and the presence of antibodies against the pleiotropic protein annexin A2 (AnxA2) in the sera of some patients after surgical resection of PDA is associated with longer recurrence-free survival. By knocking out AnxA2 in a transgenic mouse model of PDA (KPC) that recapitulates the progression of human PDA from premalignancy to metastatic disease, we found that AnxA2 promoted metastases in vivo. The expression of AnxA2 promoted the secretion of Sema3D from PDA cells, which coimmunoprecipitated with the co-receptor plexin D1 (PlxnD1) on PDA cells. Mouse PDA cells in which SEMA3D was knocked down or ANXA2-null PDA cells exhibited decreased invasive and metastatic potential in culture and in mice. However, restoring Sema3D in AnxA2-null cells did not entirely rescue metastatic behavior in culture and in vivo, suggesting that AnxA2 mediates additional prometastatic mechanisms. Patients with primary PDA tumors that have abundant Sema3D have widely metastatic disease and decreased survival compared to patients with tumors that have relatively low Sema3D abundance. Thus, AnxA2 and Sema3D may be new therapeutic targets and prognostic markers of metastatic PDA.


Sujet(s)
Annexine A2/génétique , Carcinome du canal pancréatique/génétique , Tumeurs du pancréas/génétique , Sémaphorines/génétique , Transduction du signal/génétique , Animaux , Annexine A2/métabolisme , Communication autocrine/génétique , Technique de Western , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Femelle , Analyse de profil d'expression de gènes/méthodes , Régulation de l'expression des gènes tumoraux , Humains , Protéines et peptides de signalisation intracellulaire , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Souris de souche-129 , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Microscopie de fluorescence/classification , Métastase tumorale , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Liaison aux protéines , Interférence par ARN , RT-PCR , Sémaphorines/métabolisme , Analyse de survie , Cellules cancéreuses en culture , Tumeurs du pancréas
12.
J Immunother ; 38(1): 1-11, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25415283

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDA) has a poor prognosis due to late detection and resistance to conventional therapies. Published studies show that the PDA tumor microenvironment is predominantly infiltrated with immune suppressive cells and signals that if altered, would allow effective immunotherapy. However, single-agent checkpoint inhibitors including agents that alter immune suppressive signals in other human cancers such as cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed death 1 (PD-1), and its ligand PD-L1, have failed to demonstrate objective responses when given as single agents to PDA patients. We recently reported that inhibition of the CTLA-4 pathway when given together with a T cell inducing vaccine gives objective responses in metastatic PDA patients. In this study, we evaluated blockade of the PD-1/PD-L1 pathway. We found that PD-L1 is weakly expressed at a low frequency in untreated human and murine PDAs but treatment with a granulocyte macrophage colony-stimulating factor secreting PDA vaccine (GVAX) significantly upregulates PD-L1 membranous expression after treatment of tumor-bearing mice. In addition, combination therapy with vaccine and PD-1 antibody blockade improved murine survival compared with PD-1 antibody monotherapy or GVAX therapy alone. Furthermore, PD-1 blockade increased effector CD8 T lymphocytes and tumor-specific interferon-γ production of CD8 T cells in the tumor microenvironment. Immunosuppressive pathways, including regulatory T cells and CTLA-4 expression on T cells were overcome by the addition of vaccine and low-dose cyclophosphamide to PD-1 blockade. Collectively, our study supports combining PD-1 or PD-L1 antibody therapy with a T cell inducing agent for PDA treatment.


Sujet(s)
Antigène CD274/antagonistes et inhibiteurs , Vaccins anticancéreux/immunologie , Carcinome du canal pancréatique/immunologie , Tumeurs du pancréas/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Animaux , Test ELISA , Femelle , Cytométrie en flux , Humains , Immunohistochimie , Immunothérapie/méthodes , Lymphocytes TIL/immunologie , Souris , Souris de lignée C57BL
13.
Gastroenterology ; 146(7): 1784-94.e6, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24607504

RÉSUMÉ

BACKGROUND & AIMS: Premalignant lesions and early stage tumors contain immunosuppressive microenvironments that create barriers for cancer vaccines. Kras(G12D/+);Trp53(R172H/+);Pdx-1-Cre (KPC) mice, which express an activated form of Kras in pancreatic tissues, develop pancreatic intraepithelial neoplasms (PanIN) that progress to pancreatic ductal adenocarcinoma (PDA). We used these mice to study immune suppression in PDA. METHODS: We immunized KPC and Kras(G12D/+);Pdx-1-Cre mice with attenuated intracellular Listeria monocytogenes (which induces CD4(+) and CD8(+) T-cell immunity) engineered to express Kras(G12D) (LM-Kras). The vaccine was given alone or in sequence with an anti-CD25 antibody (PC61) and cyclophosphamide to deplete T-regulatory (Treg) cells. Survival times were measured; pancreatic and spleen tissues were collected and analyzed by histologic, flow cytometry, and immunohistochemical analyses. RESULTS: Interferon γ-mediated, CD8(+) T-cell responses were observed in KPC and Kras(G12D/+);Pdx-1-Cre mice given LM-Kras, but not in unvaccinated mice. Administration of LM-Kras to KPC mice 4-6 weeks old (with early stage PanINs), depleted of Treg cells, significantly prolonged survival and reduced PanIN progression (median survival, 265 days), compared with unvaccinated mice (median survival, 150 days; P = .002), mice given only LM-Kras (median survival, 150 days; P = .050), and unvaccinated mice depleted of Treg cells (median survival, 170 days; P = .048). In 8- to 12-week-old mice (with late-stage PanINs), LM-Kras, alone or in combination with Treg cell depletion, did not increase survival time or slow PanIN progression. The combination of LM-Kras and Treg cell depletion reduced numbers of Foxp3(+)CD4(+) T cells in pancreatic lymph nodes, increased numbers of CD4(+) T cells that secrete interleukin 17 and interferon γ, and caused CD11b(+)Gr1(+) cells in the pancreas to acquire an immunostimulatory phenotype. CONCLUSIONS: Immunization of KPC mice with Listeria monocytogenes engineered to express Kras(G12D), along with depletion of Treg cells, reduces progression of early stage, but not late-stage, PanINs. This approach increases infiltration of the lesion with inflammatory cells. It might be possible to design immunotherapies against premalignant pancreatic lesions to slow or prevent progression to PDA.


Sujet(s)
Vaccins anticancéreux/usage thérapeutique , Épithélioma in situ/traitement médicamenteux , Carcinome du canal pancréatique/traitement médicamenteux , Listeria monocytogenes/immunologie , Tumeurs du pancréas/traitement médicamenteux , Lymphocytes T régulateurs/immunologie , Animaux , Anticorps monoclonaux/pharmacologie , Antigènes CD11b/métabolisme , Vaccins anticancéreux/immunologie , Épithélioma in situ/génétique , Épithélioma in situ/immunologie , Épithélioma in situ/métabolisme , Épithélioma in situ/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Cyclophosphamide/pharmacologie , Modèles animaux de maladie humaine , Évolution de la maladie , Facteurs de transcription Forkhead/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Humains , Médiateurs de l'inflammation/métabolisme , Integrases/génétique , Integrases/métabolisme , Interféron gamma/métabolisme , Interleukine-17/métabolisme , Listeria monocytogenes/génétique , Listeria monocytogenes/métabolisme , Souris , Souris de souche-129 , Souris de lignée C57BL , Souris transgéniques , Mutation , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Récepteurs aux chimiokines/métabolisme , Lymphocytes T régulateurs/métabolisme , Facteurs temps , Transactivateurs/génétique , Transactivateurs/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
14.
Hum Mol Genet ; 22(20): 4074-83, 2013 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-23727836

RÉSUMÉ

Spinal muscular atrophy (SMA) is caused by mutations of the survival motor neuron 1 (SMN1) gene, retention of the survival motor neuron 2 (SMN2) gene and insufficient expression of full-length survival motor neuron (SMN) protein. Quinazolines increase SMN2 promoter activity and inhibit the ribonucleic acid scavenger enzyme DcpS. The quinazoline derivative RG3039 has advanced to early phase clinical trials. In preparation for efficacy studies in SMA patients, we investigated the effects of RG3039 in severe SMA mice. Here, we show that RG3039 distributed to central nervous system tissues where it robustly inhibited DcpS enzyme activity, but minimally activated SMN expression or the assembly of small nuclear ribonucleoproteins. Nonetheless, treated SMA mice showed a dose-dependent increase in survival, weight and motor function. This was associated with improved motor neuron somal and neuromuscular junction synaptic innervation and function and increased muscle size. RG3039 also enhanced survival of conditional SMA mice in which SMN had been genetically restored to motor neurons. As this systemically delivered drug may have therapeutic benefits that extend beyond motor neurons, it could act additively with SMN-restoring therapies delivered directly to the central nervous system such as antisense oligonucleotides or gene therapy.


Sujet(s)
Endoribonucleases/antagonistes et inhibiteurs , Motoneurones/effets des médicaments et des substances chimiques , Amyotrophie spinale/physiopathologie , Quinazolines/pharmacologie , Petites ribonucléoprotéines nucléaires/métabolisme , Protéine-1 de survie du motoneurone/métabolisme , Animaux , Système nerveux central/effets des médicaments et des substances chimiques , Système nerveux central/métabolisme , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Évaluation préclinique de médicament , Humains , Souris , Souris transgéniques , Motoneurones/physiologie , Muscles/effets des médicaments et des substances chimiques , Muscles/métabolisme , Amyotrophie spinale/traitement médicamenteux , Amyotrophie spinale/génétique , Amyotrophie spinale/métabolisme , Jonction neuromusculaire/effets des médicaments et des substances chimiques , Jonction neuromusculaire/physiologie , Quinazolines/administration et posologie , Quinazolines/pharmacocinétique , Protéine-2 de survie du motoneurone/génétique , Protéine-2 de survie du motoneurone/métabolisme , Transmission synaptique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...