Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Mol Cancer Ther ; 21(3): 427-439, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34965960

ABSTRACT

Targeting the programmed death 1/programmed death ligand 1 (PD-1/PD-L1) pathway with immunotherapy has revolutionized the treatment of many cancers. Somatic tumor mutational burden (TMB) and T-cell-inflamed gene expression profile (GEP) are clinically validated pan-tumor genomic biomarkers that can predict responsiveness to anti-PD-1/PD-L1 monotherapy in many tumor types. We analyzed the association between these biomarkers and the efficacy of PD-1 inhibitor in 11 commonly used preclinical syngeneic tumor mouse models using murinized rat anti-mouse PD-1 DX400 antibody muDX400, a surrogate for pembrolizumab. Response to muDX400 treatment was broadly classified into three categories: highly responsive, partially responsive, and intrinsically resistant to therapy. Molecular and cellular profiling validated differences in immune cell infiltration and activation in the tumor microenvironment of muDX400-responsive tumors. Baseline and on-treatment genomic analysis showed an association between TMB, murine T-cell-inflamed gene expression profile (murine-GEP), and response to muDX400 treatment. We extended our analysis to investigate a canonical set of cancer and immune biology-related gene signatures, including signatures of angiogenesis, myeloid-derived suppressor cells, and stromal/epithelial-to-mesenchymal transition/TGFß biology previously shown to be inversely associated with the clinical efficacy of immune checkpoint blockade. Finally, we evaluated the association between murine-GEP and preclinical efficacy with standard-of-care chemotherapy or antiangiogenic agents that previously demonstrated promising clinical activity, in combination with muDX400. Our profiling studies begin to elucidate the underlying biological mechanisms of response and resistance to PD-1/PD-L1 blockade represented by these models, thereby providing insight into which models are most appropriate for the evaluation of orthogonal combination strategies.


Subject(s)
B7-H1 Antigen , Immunotherapy , Neoplasms , Programmed Cell Death 1 Receptor , Animals , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/genetics , Cell Line, Tumor , Disease Models, Animal , Humans , Immune Checkpoint Inhibitors , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment
2.
Front Immunol ; 12: 752348, 2021.
Article in English | MEDLINE | ID: mdl-34912335

ABSTRACT

Programmed cell death-1 (PD-1) blockade has a profound effect on the ability of the immune system to eliminate tumors, but many questions remain about the cell types involved and the underlying mechanisms of immune activation. To shed some light on this, the cellular and molecular events following inhibition of PD-1 signaling was investigated in the MC-38 colon carcinoma model using constitutive (PD-1 KO) and conditional (PD1cKO) mice and in wild-type mice treated with PD-1 antibody. The impact on both tumor growth and the development of tumor immunity was assessed. In the PD-1cKO mice, a complete deletion of Pdcd1 in tumor-infiltrating T cells (TILs) after tamoxifen treatment led to the inhibition of tumor growth of both small and large tumors. Extensive immune phenotypic analysis of the TILs by flow and mass cytometry identified 20-different T cell subsets of which specifically 5-CD8 positive ones expanded in all three models after PD-1 blockade. All five subsets expressed granzyme B and interferon gamma (IFNγ). Gene expression analysis of the tumor further supported the phenotypic analysis in both PD-1cKO- and PD-1 Ab-treated mice and showed an upregulation of pathways related to CD4 and CD8 T-cell activation, enhanced signaling through costimulatory molecules and IFNγ, and non-T-cell processes. Altogether, using PD-1cKO mice, we define the intrinsic nature of PD-1 suppression of CD8 T-cell responses in tumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/drug effects , Female , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency
3.
J Med Chem ; 64(8): 5137-5156, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33797901

ABSTRACT

The approvals of idelalisib and duvelisib have validated PI3Kδ inhibitors for the treatment for hematological malignancies driven by the PI3K/AKT pathway. Our program led to the identification of structurally distinct heterocycloalkyl purine inhibitors with excellent isoform and kinome selectivity; however, they had high projected human doses. Improved ligand contacts gave potency enhancements, while replacement of metabolic liabilities led to extended half-lives in preclinical species, affording PI3Kδ inhibitors with low once-daily predicted human doses. Treatment of C57BL/6-Foxp3-GDL reporter mice with 30 and 100 mg/kg/day of 3c (MSD-496486311) led to a 70% reduction in Foxp3-expressing regulatory T cells as observed through bioluminescence imaging with luciferin, consistent with the role of PI3K/AKT signaling in Treg cell proliferation. As a model for allergic rhinitis and asthma, treatment of ovalbumin-challenged Brown Norway rats with 0.3 to 30 mg/kg/day of 3c gave a dose-dependent reduction in pulmonary bronchoalveolar lavage inflammation eosinophil cell count.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/chemistry , Immunologic Factors/chemistry , Pyrrolidines/chemistry , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Binding Sites , Class I Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Dogs , Half-Life , Humans , Immunologic Factors/metabolism , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Lectins, C-Type/metabolism , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Proto-Oncogene Proteins c-akt/metabolism , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Rats , Rats, Wistar , Rhinitis, Allergic/drug therapy , Signal Transduction/drug effects , Structure-Activity Relationship
4.
Future Oncol ; 17(6): 637-648, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33300372

ABSTRACT

Tumor progression and immune evasion result from multiple oncogenic and immunosuppressive signals within the tumor microenvironment. The combined blockade of VEGF and inhibitory immune checkpoint signaling has been shown to enhance immune activation and tumor destruction in preclinical models. The LEAP clinical trial program is evaluating the safety and efficacy of lenvatinib (a multikinase inhibitor) plus pembrolizumab (a PD-1 inhibitor) across several solid tumor types. Preliminary results from ongoing trials demonstrate robust antitumor activity and durable responses across diverse tumor types with a manageable safety profile. Thus, lenvatinib plus pembrolizumab is anticipated to be an important potential new regimen for several solid cancers that currently have limited therapeutic options. Clinical trial registration: NCT03884101, NCT03713593, NCT03820986, NCT03776136, NCT03797326, NCT03829319, NCT03829332, NCT03976375, NCT04428151, NCT04199104, NCT03898180, NCT04246177 (ClinicalTrials.gov).


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Phenylurea Compounds/administration & dosage , Quinolines/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Clinical Trials as Topic , Disease Progression , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Neoplasm Staging , Neoplasms/diagnosis , Neoplasms/mortality , Neoplasms/pathology , Phenylurea Compounds/adverse effects , Progression-Free Survival , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Quinolines/adverse effects
6.
Front Immunol ; 11: 573405, 2020.
Article in English | MEDLINE | ID: mdl-33117369

ABSTRACT

The molecule "T cell immunoreceptor with immunoglobulin and ITIM domain," or TIGIT, has recently received much attention as a promising target in the treatment of various malignancies. In spite of the quick progression of anti-TIGIT antibodies into clinical testing both as monotherapy and in combination with programmed cell death-1 (PD-1)-directed immune checkpoint blockade, the molecular mechanism behind the observed therapeutic benefits remains poorly understood. Here we demonstrate, using mouse tumor models, that TIGIT blocking antibodies with functional Fc binding potential induce effective anti-tumor response. Our observations reveal that the anti-TIGIT therapeutic effect is not achieved by depletion of intratumoral regulatory T cells (Treg) or any cell population expressing TIGIT, but instead is mediated by possible "reverse activating signals" through FcγRs on myeloid cells, inducing expression of various mediators such as cytokines and chemokines. Furthermore, we discovered an induction of a robust and persistent granzyme B and perforin response, distinct from a predominantly interferon-γ (IFN-γ)-driven anti-PD-1 blockade. Our observations, for the first time, provide the basis for a mechanistic hypothesis involving the requirement of a functional Fc domain of anti-TIGIT monoclonal antibodies, of which various isotypes are currently under intense clinical investigation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Colonic Neoplasms/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Myeloid Cells/drug effects , Receptors, IgG/metabolism , Receptors, Immunologic/metabolism , Animals , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Granzymes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Signal Transduction , Tumor Burden/drug effects , Tumor Microenvironment
7.
JAMA Netw Open ; 3(2): e1920833, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32049290

ABSTRACT

Importance: Because cancer drugs given in combination have the potential for increased tumor-cell killing, finding the best combination partners for programmed cell death 1 (PD-1) checkpoint inhibitors could improve clinical outcomes for patients with cancer. Objective: To identify optimal strategies for combining PD-1 immune checkpoint inhibitors with other cancer therapies. Design, Setting, and Participants: This cross-sectional study compiled 319 results from 98 clinical trials testing PD-1 pathway inhibitors alone or in combination with other agents among 24 915 patients with metastatic cancer. All clinical trials had a primary completion date before September 16, 2018. Data analysis was conducted from November 2018 to August 2019. Exposures: Patients with metastatic cancer were treated with PD-1 immune checkpoint inhibitors alone or with other cancer therapies. Main Outcomes and Measures: Clinical activity was measured as objective response rates (ORRs). Combination measures included fold change from monotherapy to combination ORR, comparison of observed combination ORRs with estimated combination ORRs based on independent additivity, and a computational model to assess clinical synergy. To assess whether the ORRs of various combinations may be greater than the independent contribution of each agent, a Bliss independent activity model was used to analyze observed combination ORRs, and a Z score, measuring the difference between observed and calculated ORRs, was generated. Results: In 319 results from 98 clinical trials among 24 915 patients, ORRs for monotherapy were compared with combination data by indication and line of therapy, demonstrating an increased ORR in 105 of 127 results (82.7%) where ORRs were available for both PD-1 pathway inhibitor monotherapy and combination therapy. A few combinations showed increases above the Bliss-estimated activity, possibly identifying limited clinical synergy. The mean (SD) Z score for all trials was 0.0430 (0.0243). The mean (SD) Z score was 0.0923 (0.0628) for platinum chemotherapy regimen combinations, 0.0547 (0.0821) for vascular endothelial growth factor or vascular endothelial growth factor receptor tyrosine kinase inhibitor combinations, 0.0893 (0.086) for indoleamine 2,3-dioxygenase inhibitor combinations, and 0.0558 (0.0849) for cytotoxic T-lymphocyte-associated protein 4 inhibitor combinations. Conclusions and Relevance: In this cross-sectional study, most combination trials showed the expected benefit of combining 2 active anticancer agents, but few combination trials showed clinical synergy according to the Bliss independent activity model.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy/methods , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Clinical Trials as Topic , Cross-Sectional Studies , Humans , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
8.
Immunohorizons ; 3(7): 294-305, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31356159

ABSTRACT

Bromodomain and extraterminal domain (BET) proteins help direct the differentiation of helper T cell subsets, but their role in activated T cell function has not been described in detail. In this study, we investigate various consequences of epigenetic perturbation in human T lymphocytes using MK-8628, a potent and highly selective inhibitor of BET proteins. MK-8628 reduces the expression of canonical transcripts directing the proliferation, activation, and effector function of T lymphocytes. Treatment with MK-8628 abolishes the expression of key cyclins required for cell cycle progression and induces G1 cell cycle arrest in TCR-activated lymphocytes. This antiproliferative phenotype partially results from T lymphocyte apoptosis, which is exacerbated by MK-8628. In naive and memory T cell subsets, MK-8628 antagonizes T cell activation and suppresses polyfunctional cytokine production. Collectively, our results describe potent immunosuppressive effects of BET inhibition on human T cell biology. These results have important implications for immune modulatory targeting of BET proteins in the settings of T cell-driven autoimmune inflammation.


Subject(s)
Acetanilides/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Immunosuppressive Agents/pharmacology , Proteins/antagonists & inhibitors , Apoptosis/drug effects , Cell Proliferation/drug effects , Cytokines/genetics , Cytokines/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression/drug effects , Glycolysis/drug effects , Healthy Volunteers , Humans , Lymphocyte Activation/drug effects , Signal Transduction/drug effects
9.
J Clin Invest ; 128(2): 644-654, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29337311

ABSTRACT

Blockade of the checkpoint inhibitor programmed death 1 (PD1) has demonstrated remarkable success in the clinic for the treatment of cancer; however, a majority of tumors are resistant to anti-PD1 monotherapy. Numerous ongoing clinical combination therapy studies will likely reveal additional therapeutics that complement anti-PD1 blockade. Recent studies found that immunogenic cell death (ICD) improves T cell responses against different tumors, thus indicating that ICD may further augment antitumor immunity elicited by anti-PD1. Here, we observed antitumor activity following combinatorial therapy with anti-PD1 Ab and the cyclin-dependent kinase inhibitor dinaciclib in immunocompetent mouse tumor models. Dinaciclib induced a type I IFN gene signature within the tumor, leading us to hypothesize that dinaciclib potentiates the effects of anti-PD1 by eliciting ICD. Indeed, tumor cells treated with dinaciclib showed the hallmarks of ICD including surface calreticulin expression and release of high mobility group box 1 (HMGB1) and ATP. Mice treated with both anti-PD1 and dinaciclib showed increased T cell infiltration and DC activation within the tumor, indicating that this combination improves the overall quality of the immune response generated. These findings identify a potential mechanism for the observed benefit of combining dinaciclib and anti-PD1, in which dinaciclib induces ICD, thereby converting the tumor cell into an endogenous vaccine and boosting the effects of anti-PD1.


Subject(s)
Adenocarcinoma/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyridinium Compounds/pharmacology , Adenosine Triphosphate/chemistry , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , CD8-Positive T-Lymphocytes/cytology , Cell Death/drug effects , Cell Line, Tumor , Cyclic N-Oxides , Cytokines/metabolism , Dendritic Cells/cytology , Drug Synergism , Female , HMGB1 Protein/metabolism , Immune System/drug effects , Immunotherapy , Indolizines , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Phagocytosis , Programmed Cell Death 1 Receptor/metabolism , Protein Kinase Inhibitors/pharmacology
10.
Cancer Res ; 77(16): 4378-4388, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28611044

ABSTRACT

GITR is a T-cell costimulatory receptor that enhances cellular and humoral immunity. The agonist anti-mouse GITR antibody DTA-1 has demonstrated efficacy in murine models of cancer primarily by attenuation of Treg-mediated immune suppression, but the translatability to human GITR biology has not been fully explored. Here, we report the potential utility of MK-4166, a humanized GITR mAb selected to bind to an epitope analogous to the DTA-1 epitope, which enhances the proliferation of both naïve and tumor-infiltrating T lymphocytes (TIL). We also investigated the role of GITR agonism in human antitumor immune responses and report here the preclinical characterization and toxicity assessment of MK-4166, which is currently being evaluated in a phase I clinical study. Expression of human GITR was comparable with that of mouse GITR in tumor-infiltrating Tregs despite being drastically lower in other human TILs and in many human peripheral blood populations. MK-4166 decreased induction and suppressive effects of Tregsin vitro In human TIL cultures, MK-4166 induced phosphorylation of NFκB and increased expression of dual specificity phosphatase 6 (DUSP6), indicating that MK-4166 activated downstream NFκB and Erk signaling pathways. Furthermore, MK-4166 downregulated FOXP3 mRNA in human tumor infiltrating Tregs, suggesting that, in addition to enhancing the activation of TILs, MK-4166 may attenuate the Treg-mediated suppressive tumor microenvironment. Cancer Res; 77(16); 4378-88. ©2017 AACR.


Subject(s)
Antibodies/pharmacology , Glucocorticoid-Induced TNFR-Related Protein/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/immunology , Cell Line, Tumor , Female , Glucocorticoid-Induced TNFR-Related Protein/agonists , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Tumor Microenvironment
11.
Tempo psicanál ; 49(1): 37-61, jun. 2017.
Article in Portuguese | LILACS | ID: biblio-904162

ABSTRACT

A direção atual de se enxergar manifestações psíquicas como sendo, primordialmente, manifestações biológicas, tem suscitado críticas à psicanálise. Esta última é muitas vezes vista como um tratamento de menor importância quando comparada a terapias medicamentosas e comportamentais. Psicanalistas respondem apontando para a necessidade de se garantir um lugar para a singularidade de cada sujeito, em vez de uma normatização coletiva de subjetividades. Freud, embora sustentasse um projeto de ciência, afastou-se da medicina de sua época ao ver sintomas que não eram explicáveis pela biologia. Hoje, enquanto alguns defendem a atualidade das ideias de Freud, outros anunciam seu fim, argumentando que o próprio Freud cogitou que futuras evoluções no saber sobre o cérebro poderiam tornas suas ideias obsoletas. Este artigo apresenta uma reflexão sobre qual o possível lugar da psicanálise diante dos avanços da biologia atual. Entre autores a favor de uma articulação com as neurociências e autores que veem esse diálogo como um contrassenso, abordamos alguns achados neurocientíficos que se alinham às ideias de Freud; o contexto cultural que faz com que explicações fisicalistas sejam privilegiadas; a dificuldade de se entender o fenômeno mental como sendo restrito à atividade cerebral; e os pressupostos técnicos e terapêuticos da psicanálise. Ao final, refletimos sobre o que poderia ser um diálogo que respeite as especificidades de ambos os campos.


The current direction of seeing psychic manifestations as being primarily biological manifestations has sparked criticism towards psychoanalysis. The latter is often seen as a treatment of minor importance when compared to drug and behavioral therapies. Psychoanalysts respond pointing out the need to secure a place for the uniqueness of each individual rather than a collective standardization of subjectivities. Even though Freud pursued a science project, he took distance from the medicine of his time when he faced symptoms that could not be biologicaly explained. Today, as some argue the relevance of Freud's ideas, others announce their end, arguing that Freud himself wondered whether future developments in the knowledge about the brain could make his ideas obsolete. This paper presents a reflection on the role of psychoanalysis today, with all biology's advances. Among authors in favor of a connection with neurosciences and authors who see this dialogue as pointless, we address to some neuroscientific findings that align with Freud's ideas; the cultural context that privileges physicalist explanations; the difficulty in understanding the mental phenomenon as restricted to brain activity; and technical and therapeutic assumptions of psychoanalysis. Finally, we reflect on what could be a dialogue that respects the specific characteristics of both fields.

12.
Clin Cancer Res ; 22(3): 582-95, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26405194

ABSTRACT

PURPOSE: Despite significant strides in the identification and characterization of potential therapeutic targets for medulloblastoma, the role of the immune system and its interplay with the tumor microenvironment within these tumors are poorly understood. To address this, we adapted two syngeneic animal models of human Sonic Hedgehog (SHH)-driven and group 3 medulloblastoma for preclinical evaluation in immunocompetent C57BL/6 mice. EXPERIMENTAL DESIGN AND RESULTS: Multicolor flow cytometric analyses were used to phenotype and characterize immune infiltrating cells within established cerebellar tumors. We observed significantly higher percentages of dendritic cells, infiltrating lymphocytes, myeloid-derived suppressor cells, and tumor-associated macrophages in murine SHH model tumors compared with group 3 tumors. However, murine group 3 tumors had higher percentages of CD8(+) PD-1(+) T cells within the CD3 population. PD-1 blockade conferred superior antitumor efficacy in animals bearing intracranial group 3 tumors compared with SHH group tumors, indicating that immunologic differences within the tumor microenvironment can be leveraged as potential targets to mediate antitumor efficacy. Further analysis of anti-PD-1 monoclonal antibody localization revealed binding to PD-1(+) peripheral T cells, but not tumor infiltrating lymphocytes within the brain tumor microenvironment. Peripheral PD-1 blockade additionally resulted in a marked increase in CD3(+) T cells within the tumor microenvironment. CONCLUSIONS: This is the first immunologic characterization of preclinical models of molecular subtypes of medulloblastoma and demonstration that response to immune checkpoint blockade differs across subtype classification. Our findings also suggest that effective anti-PD-1 blockade does not require that systemically administered antibodies penetrate the brain tumor microenvironment.


Subject(s)
Antineoplastic Agents/pharmacology , Medulloblastoma/immunology , Medulloblastoma/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Biomarkers , Disease Models, Animal , Immunophenotyping , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Medulloblastoma/mortality , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Patched Receptors , Phenotype , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Microenvironment/genetics
13.
Blood ; 126(25): 2704-12, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26337492

ABSTRACT

Talin is an integrin adaptor, which controls integrin activity in all hematopoietic cells. How intracellular signals promote talin binding to the integrin tail leading to integrin activation is still poorly understood, especially in leukocytes. In vitro studies identified an integrin activation complex whose formation is initiated by the interaction of active, guanosine triphosphate (GTP)-bound Ras-related protein 1 (Rap1) with the adapter protein Rap1-GTP-interacting adapter molecule (RIAM) followed by the recruitment of talin to the plasma membrane. Unexpectedly, loss-of-function studies in mice have shown that the talin-activating role of RIAM is neither required for development nor for integrin activation in platelets. In this study, we show that leukocyte integrin activation critically depends on RIAM both in vitro and in vivo. RIAM deficiency results in a loss of ß2 integrin activation in multiple leukocyte populations, impaired leukocyte adhesion to inflamed vessels, and accumulation in the circulation. Surprisingly, however, the major leukocyte ß1 integrin family member, α4ß1, was only partially affected by RIAM deficiency in leukocytes. Thus, although talin is an essential, shared regulator of all integrin classes expressed by leukocytes, we report that ß2 and α4 integrins use different RIAM-dependent and -independent pathways to undergo activation by talin.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CD18 Antigens/metabolism , Chemotaxis, Leukocyte/physiology , Leukocytes/metabolism , Membrane Proteins/metabolism , Animals , Blotting, Western , Cell Adhesion/physiology , Cell Separation , Flow Cytometry , Integrin alpha4beta1/metabolism , Mice , Mice, Knockout , Talin/metabolism , rap1 GTP-Binding Proteins/metabolism
14.
Blood ; 126(25): 2695-703, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26324702

ABSTRACT

Regulation of integrins is critical for lymphocyte adhesion to endothelium and trafficking through secondary lymphoid organs. Inside-out signaling to integrins is mediated by the small GTPase Rap1. Two effectors of Rap1 regulate integrins, RapL and Rap1 interacting adaptor molecule (RIAM). Using mice conditionally deficient in both Rap1a and Rap1b and mice null for RIAM, we show that the Rap1/RIAM module is not required for T- or B-cell development but is essential for efficient adhesion to intercellular adhesion molecule (ICAM) 1 and vascular cell adhesion molecule (VCAM) 1 and for proper trafficking of lymphocytes to secondary lymphoid organs. Interestingly, in RIAM-deficient mice, whereas peripheral lymph nodes (pLNs) were depleted of both B and T cells and recirculating B cells were diminished in the bone barrow (BM), the spleen was hypercellular, albeit with a relative deficiency of marginal zone B cells. The abnormality in lymphocyte trafficking was accompanied by defective humoral immunity to T-cell-dependent antigens. Platelet function was intact in RIAM-deficient animals. These in vivo results confirm a role for RIAM in the regulation of some, but not all, leukocyte integrins and suggest that RIAM-regulated integrin activation is required for trafficking of lymphocytes from blood into pLNs and BM, where relatively high shear forces exist in high endothelial venules and sinusoids, respectively.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , B-Lymphocytes/immunology , Chemotaxis, Leukocyte/immunology , Membrane Proteins/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Adhesion/immunology , Integrins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , rap1 GTP-Binding Proteins/immunology , rap1 GTP-Binding Proteins/metabolism
15.
Sci Transl Med ; 7(279): 279ra41, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25787767

ABSTRACT

Combining immunotherapy and BRAF targeted therapy may result in improved antitumor activity with the high response rates of targeted therapy and the durability of responses with immunotherapy. However, the first clinical trial testing the combination of the BRAF inhibitor vemurafenib and the CTLA4 antibody ipilimumab was terminated early because of substantial liver toxicities. MEK [MAPK (mitogen-activated protein kinase) kinase] inhibitors can potentiate the MAPK inhibition in BRAF mutant cells while potentially alleviating the unwanted paradoxical MAPK activation in BRAF wild-type cells that lead to side effects when using BRAF inhibitors alone. However, there is the concern of MEK inhibitors being detrimental to T cell functionality. Using a mouse model of syngeneic BRAF(V600E)-driven melanoma, SM1, we tested whether addition of the MEK inhibitor trametinib would enhance the antitumor activity of combined immunotherapy with the BRAF inhibitor dabrafenib. Combination of dabrafenib and trametinib with pmel-1 adoptive cell transfer (ACT) showed complete tumor regression, increased T cell infiltration into tumors, and improved in vivo cytotoxicity. Single-agent dabrafenib increased tumor-associated macrophages and T regulatory cells (Tregs) in tumors, which decreased with the addition of trametinib. The triple combination therapy resulted in increased melanosomal antigen and major histocompatibility complex (MHC) expression and global immune-related gene up-regulation. Given the up-regulation of PD-L1 seen with dabrafenib and/or trametinib combined with antigen-specific ACT, we tested the combination of dabrafenib, trametinib, and anti-PD1 therapy in SM1 tumors, and observed superior antitumor effect. Our findings support the testing of triple combination therapy of BRAF and MEK inhibitors with immunotherapy in patients with BRAF(V600E) mutant metastatic melanoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy/methods , MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemistry , CTLA-4 Antigen/immunology , Cell Line, Tumor , Cell Survival , Humans , Imidazoles/chemistry , MAP Kinase Kinase Kinases/immunology , Major Histocompatibility Complex , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oximes/chemistry , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/immunology , Pyridones/chemistry , Pyrimidinones/chemistry , Skin Neoplasms/immunology
16.
PLoS One ; 8(12): e81152, 2013.
Article in English | MEDLINE | ID: mdl-24324670

ABSTRACT

Brucellosis is a worldwide distributed zoonosis that causes important economic losses to animal production. In Brazil, information on the distribution of biovars and genotypes of Brucella spp. is scarce or unavailable. This study aimed (i) to biotype and genotype 137 Brazilian cattle isolates (from 1977 to 2008) of B. abortus and (ii) to analyze their distribution. B. abortus biovars 1, 2 and 3 (subgroup 3b) were confirmed and biovars 4 and 6 were first described in Brazil. Genotyping by the panel 1 revealed two groups, one clustering around genotype 40 and another around genotype 28. Panels 2A and 2B disclosed a high diversity among Brazilian B. abortus strains. Eighty-nine genotypes were found by MLVA16. MLVA16 panel 1 and 2 showed geographic clustering of some genotypes. Biotyping and MLVA16 genotyping of Brazilian B. abortus isolates were useful to better understand the epidemiology of bovine brucellosis in the region.


Subject(s)
Brucella abortus/classification , Brucellosis/veterinary , Cattle Diseases/epidemiology , DNA, Bacterial/classification , Phylogeny , Zoonoses/epidemiology , Animals , Bacterial Typing Techniques , Brazil/epidemiology , Brucella abortus/genetics , Brucella abortus/isolation & purification , Brucellosis/epidemiology , Brucellosis/microbiology , Cattle , Cattle Diseases/microbiology , DNA, Bacterial/genetics , Genotype , Minisatellite Repeats , Zoonoses/microbiology
17.
Cancer Discov ; 3(7): 742-50, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23614898

ABSTRACT

The high frequency of activating RAS or BRAF mutations in cancer provides strong rationale for targeting the mitogen-activated protein kinase (MAPK) pathway. Selective BRAF and MAP-ERK kinase (MEK) inhibitors have shown clinical efficacy in patients with melanoma. However, the majority of responses are transient, and resistance is often associated with pathway reactivation of the extracellular signal-regulated kinase (ERK) signaling pathway. Here, we describe the identification and characterization of SCH772984, a novel and selective inhibitor of ERK1/2 that displays behaviors of both type I and type II kinase inhibitors. SCH772984 has nanomolar cellular potency in tumor cells with mutations in BRAF, NRAS, or KRAS and induces tumor regressions in xenograft models at tolerated doses. Importantly, SCH772984 effectively inhibited MAPK signaling and cell proliferation in BRAF or MEK inhibitor-resistant models as well as in tumor cells resistant to concurrent treatment with BRAF and MEK inhibitors. These data support the clinical development of ERK inhibitors for tumors refractory to MAPK inhibitors.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/genetics , MAP Kinase Kinase Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mutation , Neoplasms/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Signal Transduction/drug effects
18.
Nat Cell Biol ; 13(8): 989-95, 2011 Jul 24.
Article in English | MEDLINE | ID: mdl-21785421

ABSTRACT

During corticogenesis, pyramidal neurons (∼80% of cortical neurons) arise from the ventricular zone, pass through a multipolar stage to become bipolar and attach to radial glia, and then migrate to their proper position within the cortex. As pyramidal neurons migrate radially, they remain attached to their glial substrate as they pass through the subventricular and intermediate zones, regions rich in tangentially migrating interneurons and axon fibre tracts. We examined the role of lamellipodin (Lpd), a homologue of a key regulator of neuronal migration and polarization in Caenorhabditis elegans, in corticogenesis. Lpd depletion caused bipolar pyramidal neurons to adopt a tangential, rather than radial-glial, migration mode without affecting cell fate. Mechanistically, Lpd depletion reduced the activity of SRF, a transcription factor regulated by changes in the ratio of polymerized to unpolymerized actin. Therefore, Lpd depletion exposes a role for SRF in directing pyramidal neurons to select a radial migration pathway along glia rather than a tangential migration mode.


Subject(s)
Nerve Tissue Proteins/physiology , Neurogenesis/physiology , Pyramidal Cells/physiology , Serum Response Factor/physiology , Animals , Base Sequence , Cell Movement/physiology , Female , Gene Knockdown Techniques , Mice , Models, Neurological , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Pregnancy , RNA, Small Interfering/genetics
20.
Neuron ; 56(3): 441-55, 2007 Nov 08.
Article in English | MEDLINE | ID: mdl-17988629

ABSTRACT

Mammalian cortical development involves neuronal migration and neuritogenesis; this latter process forms the structural precursors to axons and dendrites. Elucidating the pathways that regulate the cytoskeleton to drive these processes is fundamental to our understanding of cortical development. Here we show that loss of all three murine Ena/VASP proteins, a family of actin regulatory proteins, causes neuronal ectopias, alters intralayer positioning in the cortical plate, and, surprisingly, blocks axon fiber tract formation during corticogenesis. Cortical fiber tract defects in the absence of Ena/VASP arise from a failure in neurite initiation, a prerequisite for axon formation. Neurite initiation defects in Ena/VASP-deficient neurons are preceded by a failure to form bundled actin filaments and filopodia. These findings provide insight into the regulation of neurite formation and the role of the actin cytoskeleton during cortical development.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation/genetics , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Microfilament Proteins/metabolism , Neurites/metabolism , Phosphoproteins/metabolism , Animals , Body Patterning/genetics , Cell Adhesion Molecules/genetics , Cell Movement/genetics , Cells, Cultured , Cerebral Cortex/cytology , Chimera , Female , Growth Cones/metabolism , Growth Cones/ultrastructure , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Mutation/genetics , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nervous System Malformations/physiopathology , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/metabolism , Neurites/ultrastructure , Phosphoproteins/genetics , Pseudopodia/metabolism , Pseudopodia/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...