Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Blood Adv ; 8(15): 4035-4049, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-38713894

RÉSUMÉ

ABSTRACT: Personalized cancer vaccines designed to target neoantigens represent a promising new treatment paradigm in oncology. In contrast to classical idiotype vaccines, we hypothesized that "polyvalent" vaccines could be engineered for the personalized treatment of follicular lymphoma (FL) using neoantigen discovery by combined whole-exome sequencing (WES) and RNA sequencing (RNA-seq). Fifty-eight tumor samples from 57 patients with FL underwent WES and RNA-seq. Somatic and B-cell clonotype neoantigens were predicted and filtered to identify high-quality neoantigens. B-cell clonality was determined by the alignment of B-cell receptor (BCR) CDR3 regions from RNA-seq data, grouping at the protein level, and comparison with the BCR repertoire from healthy individuals using RNA-seq data. An average of 52 somatic mutations per patient (range, 2-172) were identified, and ≥2 (median, 15) high-quality neoantigens were predicted for 56 of 58 FL samples. The predicted neoantigen peptides were composed of missense mutations (77%), indels (9%), gene fusions (3%), and BCR sequences (11%). Building off of these preclinical analyses, we initiated a pilot clinical trial using personalized neoantigen vaccination combined with PD-1 blockade in patients with relapsed or refractory FL (#NCT03121677). Synthetic long peptide vaccines targeting predicted high-quality neoantigens were successfully synthesized for and administered to all 4 patients enrolled. Initial results demonstrate feasibility, safety, and potential immunologic and clinical responses. Our study suggests that a genomics-driven personalized cancer vaccine strategy is feasible for patients with FL, and this may overcome prior challenges in the field. This trial was registered at www.ClinicalTrials.gov as #NCT03121677.


Sujet(s)
Antigènes néoplasiques , Vaccins anticancéreux , Lymphome folliculaire , Médecine de précision , Humains , Lymphome folliculaire/thérapie , Lymphome folliculaire/immunologie , Lymphome folliculaire/génétique , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Antigènes néoplasiques/immunologie , Médecine de précision/méthodes , Adulte d'âge moyen , Femelle , Mâle , Sujet âgé , Adulte , , Mutation
2.
Cancer Cell ; 42(3): 444-463.e10, 2024 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-38428410

RÉSUMÉ

Follicular lymphoma (FL) is a generally incurable malignancy that evolves from developmentally blocked germinal center (GC) B cells. To promote survival and immune escape, tumor B cells undergo significant genetic changes and extensively remodel the lymphoid microenvironment. Dynamic interactions between tumor B cells and the tumor microenvironment (TME) are hypothesized to contribute to the broad spectrum of clinical behaviors observed among FL patients. Despite the urgent need, existing clinical tools do not reliably predict disease behavior. Using a multi-modal strategy, we examined cell-intrinsic and -extrinsic factors governing progression and therapeutic outcomes in FL patients enrolled onto a prospective clinical trial. By leveraging the strengths of each platform, we identify several tumor-specific features and microenvironmental patterns enriched in individuals who experience early relapse, the most high-risk FL patients. These features include stromal desmoplasia and changes to the follicular growth pattern present 20 months before first progression and first relapse.


Sujet(s)
Lymphome folliculaire , Humains , Lymphocytes B , Lymphome folliculaire/génétique , Multi-omique , Études prospectives , Récidive , Microenvironnement tumoral , Essais cliniques comme sujet
3.
FASEB Bioadv ; 5(4): 156-170, 2023 Apr.
Article de Anglais | MEDLINE | ID: mdl-37020749

RÉSUMÉ

Lung cancer is the leading cause of cancer-related deaths worldwide. Surgery and chemoradiation are the standard of care in early stages of non-small cell lung cancer (NSCLC), while immunotherapy is the standard of care in late-stage NSCLC. The immune composition of the tumor microenvironment (TME) is recognized as an indicator for responsiveness to immunotherapy, although much remains unknown about its role in responsiveness to surgery or chemoradiation. In this pilot study, we characterized the NSCLC TME using mass cytometry (CyTOF) and bulk RNA sequencing (RNA-Seq) with deconvolution of RNA-Seq being performed by Kassandra, a recently published deconvolution tool. Stratification of patients based on the intratumoral abundance of B cells identified that the B-cell rich patient group had increased expression of CXCL13 and greater abundance of PD1+ CD8 T cells. The presence of B cells and PD1+ CD8 T cells correlated positively with the presence of intratumoral tertiary lymphoid structures (TLS). We then assessed the predictive and prognostic utility of these cell types and TLS within publicly available stage 3 and 4 lung adenocarcinoma (LUAD) RNA-Seq datasets. As previously described by others, pre-treatment expression of intratumoral 12-chemokine TLS gene signature is associated with progression free survival (PFS) in patients who receive treatment with immune checkpoint inhibitors (ICI). Notably and unexpectedly pre-treatment percentages of intratumoral B cells are associated with PFS in patients who receive surgery, chemotherapy, or radiation. Further studies to confirm these findings would allow for more effective patient selection for both ICI and non-ICI treatments.

4.
Sci Transl Med ; 15(680): eabn6758, 2023 01 25.
Article de Anglais | MEDLINE | ID: mdl-36696484

RÉSUMÉ

Severe and prolonged lymphopenia frequently occurs in patients with glioblastoma after standard chemoradiotherapy and has been associated with worse survival, but its underlying biological mechanism is not well understood. To address this, we performed a correlative study in which we collected and analyzed peripheral blood of patients with glioblastoma (n = 20) receiving chemoradiotherapy using genomic and immune monitoring technologies. RNA sequencing analysis of the peripheral blood mononuclear cells (PBMC) showed an elevated concentration of myeloid-derived suppressor cell (MDSC) regulatory genes in patients with lymphopenia when compared with patients without lymphopenia after chemoradiotherapy. Additional analysis including flow cytometry and single-cell RNA sequencing further confirmed increased numbers of circulating MDSC in patients with lymphopenia when compared with patients without lymphopenia after chemoradiotherapy. Preclinical murine models were also established and demonstrated a causal relationship between radiation-induced MDSC and systemic lymphopenia using transfusion and depletion experiments. Pharmacological inhibition of MDSC using an arginase-1 inhibitor (CB1158) or phosphodiesterase-5 inhibitor (tadalafil) during radiation therapy (RT) successfully abrogated radiation-induced lymphopenia and improved survival in the preclinical models. CB1158 and tadalafil are promising drugs in reducing radiation-induced lymphopenia in patients with glioblastoma. These results demonstrate the promise of using these classes of drugs to reduce treatment-related lymphopenia and immunosuppression.


Sujet(s)
Glioblastome , Lymphopénie , Cellules myéloïdes suppressives , Humains , Animaux , Souris , Glioblastome/complications , Glioblastome/traitement médicamenteux , Glioblastome/radiothérapie , Agranulocytes , Tadalafil , Lymphopénie/étiologie , Chimioradiothérapie/effets indésirables
5.
Cancer Cell ; 40(8): 879-894.e16, 2022 08 08.
Article de Anglais | MEDLINE | ID: mdl-35944503

RÉSUMÉ

Cellular deconvolution algorithms virtually reconstruct tissue composition by analyzing the gene expression of complex tissues. We present the decision tree machine learning algorithm, Kassandra, trained on a broad collection of >9,400 tissue and blood sorted cell RNA profiles incorporated into millions of artificial transcriptomes to accurately reconstruct the tumor microenvironment (TME). Bioinformatics correction for technical and biological variability, aberrant cancer cell expression inclusion, and accurate quantification and normalization of transcript expression increased Kassandra stability and robustness. Performance was validated on 4,000 H&E slides and 1,000 tissues by comparison with cytometric, immunohistochemical, or single-cell RNA-seq measurements. Kassandra accurately deconvolved TME elements, showing the role of these populations in tumor pathogenesis and other biological processes. Digital TME reconstruction revealed that the presence of PD-1-positive CD8+ T cells strongly correlated with immunotherapy response and increased the predictive potential of established biomarkers, indicating that Kassandra could potentially be utilized in future clinical applications.


Sujet(s)
Tumeurs , Transcriptome , Algorithmes , Lymphocytes T CD8+ , Humains , Apprentissage machine , Tumeurs/génétique , RNA-Seq , Analyse de séquence d'ARN , Microenvironnement tumoral/génétique
6.
Cell Rep ; 40(7): 111180, 2022 08 16.
Article de Anglais | MEDLINE | ID: mdl-35977503

RÉSUMÉ

Intratumor heterogeneity (ITH) represents a major challenge for anticancer therapies. An integrated, multidimensional, multiregional approach dissecting ITH of the clear cell renal cell carcinoma (ccRCC) tumor microenvironment (TME) is employed at the single-cell level with mass cytometry (CyTOF), multiplex immunofluorescence (MxIF), and single-nucleus RNA sequencing (snRNA-seq) and at the bulk level with whole-exome sequencing (WES), RNA-seq, and methylation profiling. Multiregional analyses reveal unexpected conservation of immune composition within each individual patient, with profound differences among patients, presenting patient-specific tumor immune microenvironment signatures despite underlying genetic heterogeneity from clonal evolution. Spatial proteogenomic TME analysis using MxIF identifies 14 distinct cellular neighborhoods and, conversely, demonstrated architectural heterogeneity among different tumor regions. Tumor-expressed cytokines are identified as key determinants of the TME and correlate with clinical outcome. Overall, this work signifies that spatial ITH occurs in ccRCC, which may drive clinical heterogeneity and warrants further interrogation to improve patient outcomes.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Protéogénomique , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Cytokines/génétique , Hétérogénéité génétique , Humains , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Analyse sur cellule unique , Microenvironnement tumoral/génétique
7.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-34769119

RÉSUMÉ

Human Ezrin Peptides (HEPs) are inhibitors of expression of IL-6 and other inflammatory cytokines, amplifiers of adaptive B cell and T cell immunity and enhancers of tissue repair. The mutation stable C-terminus of HIV gp120, mimics 69% of the "Hep-receptor", a zipped α-helical structure in the middle of the α domain of human ezrin protein. Synthetic peptides homologous to the Hep-receptor of ezrin of five to fourteen amino acids, activate anti-viral immunity against a wide range of viruses (HIV, HCV, herpes, HPV, influenza and other human respiratory viruses). Human Ezrin Peptide One (HEP1) TEKKRRETVEREKE (brand name Gepon, registered for human use in Russia from 2001) is a successful treatment for opportunistic infections in HIV-infected patients. That treats HEP1and prevents mucosal candidiasis, herpes zoster outbreaks and infection-induced chronic diarrhea. There are clinical publications in Russian on the successful treatments of chronic recurrent vaginal candidiasis, acute and chronic enterocolitis and dysbacteriosis, which are accompanied by normalization of the mucosal microbiome, and the decline or disappearance of inflammation. HEP1 is also an effective treatment and prevention for recurrent inflammation and ulceration in the stomach, duodenum and colon. HEP1 and RepG3 GEKKRRETVEREGG (a derivative of HEP1) have been used successfully as an inhaled spray peptide solution to treat a small number of human volunteers with mild-to-moderate COVID, resulting from SARS-CoV-2 infection, based on earlier successes in treating acute viral respiratory disease with inflammatory complications. Ezrin peptides seem to correct a dysregulation of innate immune responses to SARS-CoV-2. They are also adjuvants of B cell adaptive immunity and increase antibody titres, resulting in protection from lethal virus infection of mice. In a clinical study in Moscow, orally administered HEP1 was shown to enhance antibody-titres produced in response to hepatitis-B vaccination. These very preliminary but promising results with ezrin peptide treatment of COVID must be replicated in large-scale randomised placebo controlled clinical studies, to be verified.


Sujet(s)
Antiviraux/pharmacologie , Traitements médicamenteux de la COVID-19 , Protéines du cytosquelette/pharmacologie , Protéines du cytosquelette/usage thérapeutique , Immunité acquise/effets des médicaments et des substances chimiques , Adjuvants immunologiques/pharmacologie , Animaux , Antiviraux/usage thérapeutique , Infections à VIH/traitement médicamenteux , Humains , Souris , Infections de l'appareil respiratoire/traitement médicamenteux , Maladies virales/traitement médicamenteux
8.
J Immunother Cancer ; 9(7)2021 07.
Article de Anglais | MEDLINE | ID: mdl-34244308

RÉSUMÉ

BACKGROUND: Neoantigen (NeoAg) peptides displayed at the tumor cell surface by human leukocyte antigen molecules show exquisite tumor specificity and can elicit T cell mediated tumor rejection. However, few NeoAgs are predicted to be shared between patients, and none to date have demonstrated therapeutic value in the context of vaccination. METHODS: We report here a phase I trial of personalized NeoAg peptide vaccination (PPV) of 24 stage III/IV non-small cell lung cancer (NSCLC) patients who had previously progressed following multiple conventional therapies, including surgery, radiation, chemotherapy, and tyrosine kinase inhibitors (TKIs). Primary endpoints of the trial evaluated feasibility, tolerability, and safety of the personalized vaccination approach, and secondary trial endpoints assessed tumor-specific immune reactivity and clinical responses. Of the 16 patients with epidermal growth factor receptor (EGFR) mutations, nine continued TKI therapy concurrent with PPV and seven patients received PPV alone. RESULTS: Out of 29 patients enrolled in the trial, 24 were immunized with personalized NeoAg peptides. Aside from transient rash, fatigue and/or fever observed in three patients, no other treatment-related adverse events were observed. Median progression-free survival and overall survival of the 24 vaccinated patients were 6.0 and 8.9 months, respectively. Within 3-4 months following initiation of PPV, seven RECIST-based objective clinical responses including one complete response were observed. Notably, all seven clinical responders had EGFR-mutated tumors, including four patients that had continued TKI therapy concurrently with PPV. Immune monitoring showed that five of the seven responding patients demonstrated vaccine-induced T cell responses against EGFR NeoAg peptides. Furthermore, two highly shared EGFR mutations (L858R and T790M) were shown to be immunogenic in four of the responding patients, all of whom demonstrated increases in peripheral blood neoantigen-specific CD8+ T cell frequencies during the course of PPV. CONCLUSIONS: These results show that personalized NeoAg vaccination is feasible and safe for advanced-stage NSCLC patients. The clinical and immune responses observed following PPV suggest that EGFR mutations constitute shared, immunogenic neoantigens with promising immunotherapeutic potential for large subsets of NSCLC patients. Furthermore, PPV with concurrent EGFR inhibitor therapy was well tolerated and may have contributed to the induction of PPV-induced T cell responses.


Sujet(s)
Vaccins anticancéreux/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Sujet âgé , Sujet âgé de 80 ans ou plus , Vaccins anticancéreux/pharmacologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Récepteurs ErbB/métabolisme , Humains , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Mutation
9.
Cancer Cell ; 39(6): 845-865.e7, 2021 06 14.
Article de Anglais | MEDLINE | ID: mdl-34019806

RÉSUMÉ

The clinical use of molecular targeted therapy is rapidly evolving but has primarily focused on genomic alterations. Transcriptomic analysis offers an opportunity to dissect the complexity of tumors, including the tumor microenvironment (TME), a crucial mediator of cancer progression and therapeutic outcome. TME classification by transcriptomic analysis of >10,000 cancer patients identifies four distinct TME subtypes conserved across 20 different cancers. The TME subtypes correlate with patient response to immunotherapy in multiple cancers, with patients possessing immune-favorable TME subtypes benefiting the most from immunotherapy. Thus, the TME subtypes act as a generalized immunotherapy biomarker across many cancer types due to the inclusion of malignant and microenvironment components. A visual tool integrating transcriptomic and genomic data provides a global tumor portrait, describing the tumor framework, mutational load, immune composition, anti-tumor immunity, and immunosuppressive escape mechanisms. Integrative analyses plus visualization may aid in biomarker discovery and the personalization of therapeutic regimens.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Immunothérapie/méthodes , Tumeurs/étiologie , Tumeurs/thérapie , Microenvironnement tumoral/immunologie , Fibroblastes associés au cancer/immunologie , Fibroblastes associés au cancer/anatomopathologie , Visualisation de données , Bases de données factuelles , Analyse de profil d'expression de gènes/méthodes , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Mélanome/génétique , Mélanome/immunologie , Mélanome/anatomopathologie , Tumeurs/mortalité , Tumeurs/anatomopathologie , Médecine de précision , Tumeurs cutanées/génétique , Tumeurs cutanées/immunologie , Tumeurs cutanées/anatomopathologie , Résultat thérapeutique , Microenvironnement tumoral/génétique
10.
Clin Cancer Res ; 27(12): 3478-3490, 2021 06 15.
Article de Anglais | MEDLINE | ID: mdl-33771855

RÉSUMÉ

PURPOSE: Multiparametric MRI (mpMRI) has become an indispensable radiographic tool in diagnosing prostate cancer. However, mpMRI fails to visualize approximately 15% of clinically significant prostate cancer (csPCa). The molecular, cellular, and spatial underpinnings of such radiographic heterogeneity in csPCa are unclear. EXPERIMENTAL DESIGN: We examined tumor tissues from clinically matched patients with mpMRI-invisible and mpMRI-visible csPCa who underwent radical prostatectomy. Multiplex immunofluorescence single-cell spatial imaging and gene expression profiling were performed. Artificial intelligence-based analytic algorithms were developed to examine the tumor ecosystem and integrate with corresponding transcriptomics. RESULTS: More complex and compact epithelial tumor architectures were found in mpMRI-visible than in mpMRI-invisible prostate cancer tumors. In contrast, similar stromal patterns were detected between mpMRI-invisible prostate cancer and normal prostate tissues. Furthermore, quantification of immune cell composition and tumor-immune interactions demonstrated a lack of immune cell infiltration in the malignant but not in the adjacent nonmalignant tissue compartments, irrespective of mpMRI visibility. No significant difference in immune profiles was detected between mpMRI-visible and mpMRI-invisible prostate cancer within our patient cohort, whereas expression profiling identified a 24-gene stromal signature enriched in mpMRI-invisible prostate cancer. Prostate cancer with strong stromal signature exhibited a favorable survival outcome within The Cancer Genome Atlas prostate cancer cohort. Notably, five recurrences in the 8 mpMRI-visible patients with csPCa and no recurrence in the 8 clinically matched patients with mpMRI-invisible csPCa occurred during the 5-year follow-up post-prostatectomy. CONCLUSIONS: Our study identified distinct molecular, cellular, and structural characteristics associated with mpMRI-visible csPCa, whereas mpMRI-invisible tumors were similar to normal prostate tissue, likely contributing to mpMRI invisibility.


Sujet(s)
Imagerie par résonance magnétique multiparamétrique , Tumeurs de la prostate , Intelligence artificielle , Écosystème , Humains , Mâle , Tumeurs de la prostate/imagerie diagnostique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/chirurgie , Protéomique
11.
Sci Rep ; 9(1): 4563, 2019 03 14.
Article de Anglais | MEDLINE | ID: mdl-30872589

RÉSUMÉ

Signaling via Toll-like receptor 4 (TLR4) in macrophages constitutes an essential part of the innate immune response to bacterial infections. Detailed and quantified descriptions of TLR4 signal transduction would help to understand and exploit the first-line response of innate immune defense. To date, most mathematical modelling studies were performed on transformed cell lines. However, properties of primary macrophages differ significantly. We therefore studied TLR4-dependent activation of NF-κB transcription factor in bone marrow-derived and peritoneal primary macrophages. We demonstrate that the kinetics of NF-κB phosphorylation and nuclear translocation induced by a wide range of bacterial lipopolysaccharide (LPS) concentrations in primary macrophages is much faster than previously reported for macrophage cell lines. We used a comprehensive combination of experiments and mathematical modeling to understand the mechanisms of this rapid response. We found that elevated basal NF-κB in the nuclei of primary macrophages is a mechanism increasing native macrophage sensitivity and response speed to the infection. Such pre-activated state of macrophages accelerates the NF-κB translocation kinetics in response to low agonist concentrations. These findings enabled us to refine and construct a new model combining both NF-κB phosphorylation and translocation processes and predict the existence of a negative feedback loop inactivating phosphorylated NF-κB.


Sujet(s)
Noyau de la cellule/métabolisme , Lipopolysaccharides/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire , Cytosol/métabolisme , Activation des macrophages/immunologie , Modèles biologiques , Phosphorylation , Transport des protéines , Transduction du signal , Récepteur de type Toll-4/agonistes , Récepteur de type Toll-4/métabolisme , Ubiquitination
12.
BMC Immunol ; 19(1): 26, 2018 07 28.
Article de Anglais | MEDLINE | ID: mdl-30055563

RÉSUMÉ

BACKGROUND: Agonists of TLR3 and TLR4 are effective immunoadjuvants for different types of vaccines. The mechanisms of their immunostimulatory action differ significantly; these differences are particularly critical for immunization with non-replicating adenovirus vectors (rAds) based vaccines. Unlike traditional vaccines, rAd based vaccines are not designed to capture vaccine antigens from the external environment by antigen presenting cells (APCs), but rather they are targeted to the de novo synthesis of vaccine antigens in APCs transfected with rAd. To date, there is no clear understanding about approaches to improve the efficacy of rAd vaccinations with immunoadjuvants. In this study, we investigated the immunoadjuvant effect of TLR3 and TLR4 agonists on the level of activation of APCs during vaccination with rAds. RESULTS: We demonstrated that TLR3 and TLR4 agonists confer different effects on the molecular processes in APCs that determine the efficacy of antigen delivery and activation of antigen-specific CD4+ and CD8+ T cells. APCs activated with agonists of TLR4 were characterized by up-regulated production of target antigen mRNA and protein encoded in rAd, as well as enhanced expression of the co-activation receptors CD80, CD86 and CD40, and pro-inflammatory cytokines TNF-α, IL6 and IL12. These effects of TLR4 agonists have provided a significant increase in the number of antigen-specific CD4+ and CD8+ T cells. TLR3 agonist, on the contrary, inhibited transcription and synthesis of rAd-encoded antigens, but improved expression of CD40 and IFN-ß in APCs. The cumulative effect of TLR3 agonist have resulted in only a slight improvement in the activation of antigen-specific T cells. Also, we demonstrated that IFN-ß and TNF-α, secreted by APCs in response to TLR3 and TLR4 agonists, respectively, have an opposite effect on the transcription of the targeted gene encoded in rAd. Specifically, IFN-ß inhibited, and TNF-α stimulated the expression of target vaccine antigens in APCs. CONCLUSIONS: Our data demonstrate that agonists of TLR4 but not TLR3 merit further study as adjuvants for development of vaccines based on recombinant adenoviral vectors.


Sujet(s)
Vaccins anti-adénovirus/immunologie , Adjuvants immunologiques/pharmacologie , Présentation d'antigène , Cellules dendritiques/effets des médicaments et des substances chimiques , Récepteur de type Toll-3/agonistes , Récepteur de type Toll-4/agonistes , Animaux , Antigène CD80/immunologie , Antigène CD86/immunologie , Lymphocytes T CD4+/immunologie , Antigènes CD40/immunologie , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Vecteurs génétiques/immunologie , Immunisation , Interféron bêta/immunologie , Interleukine-12/immunologie , Interleukine-6/immunologie , Souris , Souris de lignée BALB C , Lymphocytes auxiliaires Th1/immunologie , Facteur de nécrose tumorale alpha/immunologie , Vaccins synthétiques/immunologie
13.
J Immunol ; 200(8): 2656-2669, 2018 04 15.
Article de Anglais | MEDLINE | ID: mdl-29500244

RÉSUMÉ

Dendritic cells (DCs) are well-known for their functions in orchestrating the innate and adaptive arms of immune defense. However, under certain conditions, DCs can exert tumoricidal activity. We have elucidated the mechanism of tumor suppression by TLR4-activated bone marrow-derived DCs (BMDCs) isolated from BALB/c mice. We identified that two distinct subsets of BMDCs (CD11b+CD11c+I-A/Eint and CD11b+CD11c+I-A/Ehigh) have different cytotoxic mechanisms of action. The cytotoxicity of the former subset is mediated through NO and reactive oxygen species and type I IFN (IFN-ß), whereas the latter subset acts only through IFN-ß. TLR4 agonists, LPS or pharmaceutical-grade ImmunoMax, activate CD11c+ BMDCs, which, in turn, directly kill 4T1 mouse breast cancer cells or inhibit their proliferation in an MHC-independent manner. These data define two populations of BMDCs with different mechanisms of direct cytotoxicity, as well as suggest that the I-A/Eint subset could be less susceptible to counteracting mechanisms in the tumor microenvironment and support investigation of similar subsets in human DCs.


Sujet(s)
Moelle osseuse/métabolisme , Cellules dendritiques/métabolisme , Récepteur de type Toll-4/agonistes , Animaux , Cellules de la moelle osseuse/métabolisme , Antigènes CD11c/métabolisme , Lignée cellulaire tumorale , Cellules cultivées , Femelle , Interféron bêta/métabolisme , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Microenvironnement tumoral/physiologie
14.
PLoS One ; 13(1): e0191574, 2018.
Article de Anglais | MEDLINE | ID: mdl-29377916

RÉSUMÉ

To avoid outbreaks of influenza virus epidemics and pandemics among human populations, modern medicine requires the development of new universal vaccines that are able to provide protection from a wide range of influenza A virus strains. In the course of development of a universal vaccine, it is necessary to consider that immunity must be generated even against viruses from different hosts because new human epidemic virus strains have their origins in viruses of birds and other animals. We have enriched conserved viral proteins-nucleoprotein (NP) and matrix protein 2 (M2)-by B and T-cell epitopes not only human origin but also swine and avian origin. For this purpose, we analyzed M2 and NP sequences with respect to changes in the sequences of known T and B-cell epitopes and chose conserved and evolutionarily significant epitopes. Eventually, we found consensus sequences of M2 and NP that have the maximum quantity of epitopes that are 100% coincident with them. Consensus epitope-enriched amino acid sequences of M2 and NP proteins were included in a recombinant adenoviral vector. Immunization with Ad5-tet-M2NP induced strong CD8 and CD4 T cells responses, specific to each of the encoded antigens, i.e. M2 and NP. Eight months after immunization with Ad5-tet-M2NP, high numbers of M2- and NP-responding "effector memory" CD44posCD62neg T cells were found in the mouse spleens, which revealed a long-term T cell immune memory conferred by the immunization. In all, the challenge experiments showed an extraordinarily wide-ranging efficacy of protection by the Ad5-tet-M2NP vaccine, covering 5 different heterosubtypes of influenza A virus (2 human, 2 avian and 1 swine).


Sujet(s)
Lymphocytes B/immunologie , Épitopes/immunologie , Virus de la grippe A/immunologie , Nucléoprotéines/immunologie , Lymphocytes T/immunologie , Vaccination , Protéines de la matrice virale/immunologie , Animaux , Anticorps antiviraux/biosynthèse
16.
Eur J Pharmacol ; 811: 249-259, 2017 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-28668507

RÉSUMÉ

Synthetic 14 AA peptide (Gepon) derived from the hinge region of ezrin, a protein that links cell surface molecules to intracellular actin filaments, accelerates and facilitates wound and ulcer healing in clinical applications. However, the molecular mechanisms underlying this phenomenon and involved in enhanced healing of wounds with Gepon are not yet understood. The purpose of current study was to investigate intracellular signaling pathways involved in the effect of this peptide on wild type and genetically modified (CD44 KO) NIH/3T3 embryonic mouse fibroblasts. Gepon treatment of NIH/3T3 cells resulted in morphological and biochemical changes, characteristic of differentiated fibroblasts. While treatment of NIH/3T3 cells with TGF-ß1 triggered the activation of both canonical and non-canonical signaling pathways, exposure of fibroblasts to Gepon activated only the ERK1/2 dependent pathway without modulating SMAD dependent signaling pathway. Knocking out hyaluronic acid CD44 receptor did not change Gepon or TGF-ß1 dependent activation of intracellular signaling pathways and assembling of α-SMA-positive filaments. Gepon dependent differentiation of NIH/3T3 fibroblasts is based on activation of ERK1/2 kinase, non-canonical intracellular signaling pathway. Our data suggest that the treatment of fibroblasts with Gepon triggers activation of the non-canonical (SMAD independent) intracellular signaling pathway that involves ERK1/2kinase phosphorylation. Activation of the MAPK signaling pathway and the increase in formation of α-SMA containing stress filaments induced by Gepon were independent on presence of CD44 receptor in NIH/3T3 fibroblasts. Thus, our observation designates the significance and sufficiency of MAPK pathway mediated activation of fibroblasts with Gepon for healing of erosion, ulcers and wounds.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Protéines du cytosquelette/composition chimique , Fibroblastes/cytologie , Fibroblastes/effets des médicaments et des substances chimiques , Fragments peptidiques/composition chimique , Fragments peptidiques/pharmacologie , Séquence d'acides aminés , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Collagène de type I/génétique , Fibroblastes/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Techniques de knock-out de gènes , Antigènes CD44/déficit , Antigènes CD44/génétique , Espace intracellulaire/effets des médicaments et des substances chimiques , Espace intracellulaire/métabolisme , Souris , Cellules NIH 3T3 , Protéines proto-oncogènes c-fos/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de croissance transformant alpha/métabolisme
17.
J Transl Med ; 12: 322, 2014 Nov 29.
Article de Anglais | MEDLINE | ID: mdl-25432242

RÉSUMÉ

BACKGROUND: Previously we demonstrated that the resection of primary 4T1 tumors only slightly prolongs mouse survival, but importantly, creates a "window of opportunity" with attenuated suppressor cell and increased activated T cell populations. This suggests that additional activation of the immune system by immunostimulatory agents during this period may enhance anti-tumor immunity and potentially eradicate micro-metastatic disease in this stringent model. We hypothesized that the immunostimulator Immunomax®, which is comprised of a plant-derived polysaccharide, is non-toxic in humans and stimulates immune defense during the infectious diseases treatment, may have also anti-tumor activity and be beneficial in the adjuvant setting when endogenous anti-tumor responses are present and during the "window of opportunity" in post-resection metastatic breast cancer model. Here we provide the initial report that Immunomax® demonstrates the capacity to eliminate micro-metastatic disease in the post-resection, 4T1 mouse model of breast cancer. METHODS: The efficacy of Immunomax® was evaluated by analyzing survival rate and the number of spontaneous clonogenic tumor cells in the lung homogenates of mice. The frequencies of activated NK, CD4(+) and CD8(+) cells as well as myeloid-derived suppressor cells and Treg cells were evaluated using flow cytometry. Highly purified mouse and human dendritic and NK cells were sorted and the effect of Immunomax® on activation status of these cells was assessed by flow cytometry. The property of Immunomax® as TLR-4 agonist was determined by NF-κB/SEAP reporter gene assay, WB, RT-PCR. RESULTS: Immunomax® injections significantly prolonged overall survival and cured 31% of mice. This immunostimulator activates DCs via the TLR-4, which in turn stimulates tumoricidal NK cells and in vitro, completely inhibits growth of 4T1 cells. Incubation of PBMC from healthy donors with Immunomax® activates NK cells via activation of plasmacytoid DC leading significantly higher efficacy in killing of human NK-target cells K562 compared with non-treated cells. CONCLUSION: This is the first demonstration that Immunomax® is a TLR-4 agonist and the first report of a documented role for this pharmaceutical grade immunostimulator in augmenting anti-tumor activity, suggesting that incorporation of Immunomax® into developing breast cancer therapeutic strategies may be beneficial and with less potential toxicity than checkpoint inhibitors.


Sujet(s)
Tumeurs expérimentales de la mamelle/thérapie , Métastase tumorale , Extraits de plantes/pharmacologie , Récepteur de type Toll-4/effets des médicaments et des substances chimiques , Animaux , Femelle , Lymphocytes/immunologie , Tumeurs expérimentales de la mamelle/immunologie , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris de lignée BALB C
18.
Clin Exp Metastasis ; 31(2): 185-98, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24096737

RÉSUMÉ

It is believed that primary tumor resection modulates host-tumor immune interaction, but this has not been characterized in a stringent breast cancer tumor model. This report, using the 4T1 murine mammary tumor model, characterizes for the first time the dynamic longitudinal changes in immunosuppressive and effector components of the immune system after resection of an established orthotopic primary tumor with a defined natural history of developing lung metastases. More specifically, we analyzed changes of absolute numbers and frequencies of MDSC, regulatory T cells (Treg), as well as activated CD4 and CD8 positive T cells in spleens and, in some studies, lungs of 4T1 tumor-bearing mice and mice after primary tumor resection. Importantly, using mathematical analyses we established that primary resection of an orthotopic tumor had created a "window of opportunity" with decreased tumor-associated immune suppression that existed for approximately 10 days. Although tumor resection did slightly prolong survival, it did not affect the ultimate development of metastatic disease since animals with resected tumors or intact primary tumors eventually died by day 47 and 43, respectively. This window of opportunity likely occurs in humans providing a rationale and parameters for integration and testing of immunotherapeutic strategies in this critical "window of opportunity" to combat the development of metastatic disease.


Sujet(s)
Immunothérapie , Tumeurs expérimentales de la mamelle/chirurgie , Tumeurs expérimentales de la mamelle/thérapie , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Tumeurs expérimentales de la mamelle/immunologie , Souris
19.
Arzneimittelforschung ; 60(6): 324-9, 2010.
Article de Anglais | MEDLINE | ID: mdl-20648922

RÉSUMÉ

The therapeutic agent OM-89 (Uro-Vaxom) contains lyophilized immunostimulating fractions from 18 Escherichia coil strains. It has been shown to provide protection against recurrent urinary tract infections in humans and against bacterial infections in mice. Here the immunostimulatory properties of OM-89 were investigated by in vitro and in vivo assays. In vitro the activation of murine spleen cells by the AlamarBlue assay was determined. OM-89 was effective in stimulating the metabolism of spleen cells within a concentration range of 0.625-2.5 mg/ml. The activation of murine bone marrow-derived macrophages by OM-89 was shown by the induction of NO production; OM-89 was a most effective stimulant at concentrations around 6 mg/ml. In the human system, the effect of OM-89 was tested in vitro:metabolic activity of peripheral blood lymphocytes (PBL) was stimulated starting at concentrations of approx. 250 microg/ml, and the spontaneous apoptosis of polymorphonuclear neutrophils (PMN) was reduced starting at OM-89 concentrations of approx. 100 microg/ml. Finally, in a mouse model, the in vivo protection of mice against infection with Salmonella typhimurium after the oral administration of OM-89 was tested (100 mg in a volume of 0.5 ml once a day for 10 consecutive days). The extract proved to be effective: 90% of the OM-89-treated animals survived compared to 58% of the untreated control group.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Anti-infectieux/pharmacologie , Antigènes bactériens/pharmacologie , Escherichia coli/effets des médicaments et des substances chimiques , Animaux , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/physiologie , Femelle , Macrophages/effets des médicaments et des substances chimiques , Macrophages/physiologie , Souris , Souris de lignée BALB C , Monoxyde d'azote/biosynthèse , Rate/effets des médicaments et des substances chimiques , Rate/physiologie
20.
Vaccine ; 28(8): 1987-96, 2010 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-20188254

RÉSUMÉ

The aim of this study was to evaluate the immunogenicity of NS5A protein of human hepatitis C virus (HCV) when delivered as naked DNA (NS5A DNA), or recombinant protein (rNS5A). DBA/2J mice received NS5A DNA, rNS5A, or NS5A DNA/rNS5A in different prime-boost combinations with a peptidoglycan Immunomax((R)). The weakest response was induced after rNS5A prime and NS5A DNA boost; rNS5A alone induced an immune response with a strong Th2-component; and NS5A DNA alone, a relatively weak secretion of IL-2 and IFN-gamma. The most efficient was co-injection of NS5A DNA and rNS5A, which induced a significant increase in CD4(+) and CD8(+) T-cell counts, anti-NS5A antibodies, specific T-cell proliferation, and proinflammatory cytokine production in vitro against a broad spectrum of NS5A epitopes. Administration of the mixture of adjuvanted DNA and protein immunogens can be selected as the best regimen for further preclinical HCV-vaccine trials.


Sujet(s)
Hépatite C/prévention et contrôle , Vaccins à ADN/immunologie , Vaccins contre les hépatites virales/immunologie , Protéines virales non structurales/immunologie , Adjuvants immunologiques/pharmacologie , Animaux , Antigènes viraux/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Lignée cellulaire , Prolifération cellulaire , Épitopes/immunologie , Femelle , Hépatite C/immunologie , Humains , Rappel de vaccin , Interféron gamma/immunologie , Interleukine-2/immunologie , Souris , Souris de lignée DBA , Peptidoglycane/immunologie , Protéines recombinantes/immunologie , Lymphocytes auxiliaires Th2/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE