Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 198(10): 4166-4177, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28396317

RESUMO

Myeloid cells play a key role in tumor progression and metastasis by providing nourishment and immune protection, as well as facilitating cancer invasion and seeding to distal sites. Although advances have been made in understanding the biology of these tumor-educated myeloid cells (TEMCs), their intrinsic plasticity challenges our further understanding of their biology. Indeed, in vitro experiments only mimic the in vivo setting, and current gene-knockout technologies do not allow the simultaneous, temporally controlled, and cell-specific silencing of multiple genes or pathways. In this article, we describe the 4PD nanoplatform, which allows the in vivo preferential transfection and in vivo tracking of TEMCs with the desired RNAs. This platform is based on the conjugation of CD124/IL-4Rα-targeting peptide with G5 PAMAM dendrimers as the loading surface and can convey therapeutic or experimental RNAs of interest. When injected i.v. in mice bearing CT26 colon carcinoma or B16 melanoma, the 4PD nanoparticles predominantly accumulate at the tumor site, transfecting intratumoral myeloid cells. The use of 4PD to deliver a combination of STAT3- and C/EBPß-specific short hairpin RNA or miR-142-3p confirmed the importance of these genes and microRNAs in TEMC biology and indicates that silencing of both genes is necessary to increase the efficacy of immune interventions. Thus, the 4PD nanoparticle can rapidly and cost effectively modulate and assess the in vivo function of microRNAs and mRNAs in TEMCs.


Assuntos
Dendrímeros/metabolismo , Inativação Gênica , Células Mieloides/metabolismo , Nanotecnologia/métodos , Animais , Linhagem Celular Tumoral , Neoplasias do Colo , Dendrímeros/administração & dosagem , Subunidade alfa de Receptor de Interleucina-4/imunologia , Subunidade alfa de Receptor de Interleucina-4/metabolismo , Melanoma Experimental , Camundongos , MicroRNAs , Células Mieloides/imunologia , Nanopartículas/administração & dosagem , Nanopartículas/metabolismo , Nanotecnologia/normas , Receptores de Interleucina-4/imunologia , Receptores de Interleucina-4/metabolismo
2.
Monaldi Arch Chest Dis ; 88(1): 888, 2018 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-29741076

RESUMO

Nosocomial or hospital acquired pneumonia (HAP) is an illness contracted during a hospital stay, generally with onset 48 hours or more after admission to hospital, or within 14 days of discharge from hospital. HAP is divided into subgroups: Ventilator-associated pneumonia (VAP), accounting for 86% of hospital acquired pneumonia, and stroke-associated pneumonia (SAP). The incidence of SAP in neurological intensive care units (NICUs) is 4.1-56.6%, in medical intensive care units (MICUs) it is 17-50%, in stroke units it is 3.9-44% and in rehabilitation it is 3.2-11%, whereas in intensive rehabilitation following severe cranial trauma, the reported incidence of HAP is between 3.9 and 12% of cases. The aim of this study is to evaluate the cases of HAP occurring in a continuous series of patients with severe acquired brain injury (sABI) admitted to intensive rehabilitation units. The data collected can help evaluate the growing complexity of early rehabilitation of these patients, starting from how lung infections interfere with hospital stays and rehabilitation outcomes. This prospective observational cohort study evaluates, from 01/01/2015 to 31/12/2015, for patients with sABI on first admission to intensive neurological rehabilitation, the frequency of HAP and its impact on patient outcomes and complexity of care. A total of 61 patients were enrolled: 39 males and 22 females, average age 59.5 years (17-88 yrs, SD 3.53), coming from critical care (n=52), medical units (n=5), neurosurgery (n=3) and surgical units (n=1). The aetiology of hospital admission was haemorrhagic in 36% of cases, traumatic in 36%, anoxic in 13.1%, infectious in 6.5%, ischaemic in 4.9%, and other causes in 3.2%. Among the patients, 93.44% had received antibiotic therapy in their unit of provenance, and in 61.27% of cases a multidrug resistant (MDR) bacterium was isolated. On enrolment, 26 patients presented respiratory insufficiency, 29 subjects were in oxygen therapy, and 4 under invasive mechanical ventilation. There were 54 tracheostomized patients, 33 patients with percutaneous endoscopic gastrostomy (PEG) tubes, and 23 with nasogastric (NG) tubes. In 2015, among these subjects admitted to neurological rehabilitation, the incidence of HAP was 13.1%. For these 8 pneumonia cases, it was possible to isolate the bacterium in 62.5% of cases, and the detected microorganisms were K. pneumoniae (n=2), P. aeruginosa (n=1), P. mirabilis (n=1), S. maltophilia (n=1), E. cloacae + MRSCoN (n=1). Compared to the literature data, the results of the first year of monitoring show a slight increase in HAP cases (13.1%) in severe brain injury patients on first admission to neurological rehabilitation. These preliminary results need to be further confirmed and monitored over time. The findings moreover confirm the criticality and complexity of care for these patients admitted to neurological rehabilitation units.

3.
Eur J Immunol ; 44(11): 3307-19, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25113564

RESUMO

By restraining T-cell activation and promoting Treg-cell expansion, myeloid-derived suppressor cells (MDSCs) and tolerogenic DCs can control self-reactive and antigraft effector T cells in autoimmunity and transplantation. Their therapeutic use and characterization, however, is limited by their scarce availability in the peripheral blood of tumor-free donors. In the present study, we describe and characterize a novel population of human myeloid suppressor cells, named fibrocytic MDSC, which are differentiated from umbilical cord blood precursors by 4-day culture with FDA-approved cytokines (recombinant human-GM-CSF and recombinant human-G-CSF). This MDSC subset, characterized by the expression of MDSC-, DC-, and fibrocyte-associated markers, promotes Treg-cell expansion and induces normoglycemia in a xenogeneic mouse model of Type 1 diabetes. In order to exert their protolerogenic function, fibrocytic MDSCs require direct contact with activated T cells, which leads to the production and secretion of IDO. This new myeloid subset may have an important role in the in vitro and in vivo production of Treg cells for the treatment of autoimmune diseases, and in either the prevention or control of allograft rejection.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Ativação Linfocitária/imunologia , Células Mieloides/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular , Proliferação de Células , Diabetes Mellitus Tipo 1/imunologia , Feminino , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Rejeição de Enxerto/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células HEK293 , Humanos , Tolerância Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Mieloides/citologia , Proteínas Recombinantes/farmacologia , Linfócitos T Reguladores/transplante
4.
J Infect Dis ; 208(11): 1914-22, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23901083

RESUMO

BACKGROUND: Amphotericin B (AmB), the most effective drug against leishmaniasis, has serious toxicity. As Leishmania species are obligate intracellular parasites of antigen presenting cells (APC), an immunopotentiating APC-specific AmB nanocarrier would be ideally suited to reduce the drug dosage and regimen requirements in leishmaniasis treatment. Here, we report a nanocarrier that results in effective treatment shortening of cutaneous leishmaniasis in a mouse model, while also enhancing L. major specific T-cell immune responses in the infected host. METHODS: We used a Pan-DR-binding epitope (PADRE)-derivatized-dendrimer (PDD), complexed with liposomal amphotericin B (LAmB) in an L. major mouse model and analyzed the therapeutic efficacy of low-dose PDD/LAmB vs full dose LAmB. RESULTS: PDD was shown to escort LAmB to APCs in vivo, enhanced the drug efficacy by 83% and drug APC targeting by 10-fold and significantly reduced parasite burden and toxicity. Fortuitously, the PDD immunopotentiating effect significantly enhanced parasite-specific T-cell responses in immunocompetent infected mice. CONCLUSIONS: PDD reduced the effective dose and toxicity of LAmB and resulted in elicitation of strong parasite specific T-cell responses. A reduced effective therapeutic dose was achieved by selective LAmB delivery to APC, bypassing bystander cells, reducing toxicity and inducing antiparasite immunity.


Assuntos
Anfotericina B/administração & dosagem , Antiprotozoários/administração & dosagem , Dendrímeros/administração & dosagem , Leishmania major/efeitos dos fármacos , Leishmaniose Cutânea/tratamento farmacológico , Vacinas Antimaláricas/administração & dosagem , Imunidade Adaptativa , Anfotericina B/toxicidade , Animais , Células Apresentadoras de Antígenos/imunologia , Antiprotozoários/toxicidade , Modelos Animais de Doenças , Portadores de Fármacos , Epitopos , Feminino , Injeções Intraperitoneais , Leishmania major/imunologia , Vacinas contra Leishmaniose , Leishmaniose Cutânea/imunologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Nanopartículas
5.
Comput Oper Res ; 40(11): 2625-2631, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24187428

RESUMO

We consider an uncertain variant of the knapsack problem in which the weight of the items is not exactly known in advance, but belongs to a given interval, and an upper bound is imposed on the number of items whose weight differs from the expected one. For this problem, we provide a dynamic programming algorithm and present techniques aimed at reducing its space and time complexities. Finally, we computationally compare the performances of the proposed algorithm with those of different exact algorithms presented so far in the literature for robust optimization problems.

6.
Cancer Res ; 83(23): 3886-3900, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37602821

RESUMO

Emerging evidence suggests that not only the frequency and composition of tumor-infiltrating leukocytes but also their spatial organization might be a major determinant of tumor progression and response to therapy. Therefore, mapping and analyzing the fine tumor immune architecture could potentially provide insights for predicting cancer prognosis. Here, we performed an explorative, prospective clinical study to assess whether structures within the tumor microenvironment can predict recurrence after salvage surgery in head and neck squamous cell carcinoma (HNSCC). The major immune subsets were measured using flow cytometry and co-detection by indexing (CODEX) multiparametric imaging. Flow cytometry underestimated the number of PMN-MDSCs and neutrophils in the tumor and overestimated the tumor-infiltrating lymphocyte frequency. An ad hoc computational framework was used to identify and analyze discrete cellular neighborhoods. A high frequency of tertiary lymphoid structures composed of CD31highCD38high plasma cells was associated with reduced recurrence after surgery in HNSCC. These data support the notion that the structural architecture of the tumor immune microenvironment plays an essential role in tumor progression and indicates that type 1 tertiary lymphoid structures and long-lived CD31highCD38high plasma cells are associated with good prognosis in HNSCC. SIGNIFICANCE: Imaging the spatial tumor immune microenvironment and evaluating the presence of type 1 tertiary lymphoid structures enables prediction of recurrence after surgery in patients with head and neck squamous cell carcinoma.


Assuntos
Neoplasias de Cabeça e Pescoço , Estruturas Linfoides Terciárias , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Neoplasias de Cabeça e Pescoço/cirurgia , Microambiente Tumoral , Estudos Prospectivos , Prognóstico
7.
J Exp Med ; 203(12): 2691-702, 2006 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-17101732

RESUMO

Phosphodiesterase-5 (PDE5) inhibitors (sildenafil, tadalafil, and vardenafil) are agents currently in clinical use for nonmalignant conditions. We report the use of PDE5 inhibitors as modulators of the antitumor immune response. In several mouse tumor models, PDE5 inhibition reverses tumor-induced immunosuppressive mechanisms and enables a measurable antitumor immune response to be generated that substantially delays tumor progression. In particular, sildenafil, down-regulates arginase 1 and nitric oxide synthase-2 expression, thereby reducing the suppressive machinery of CD11b+/Gr-1+ myeloid-derived suppressor cells (MDSCs) recruited by growing tumors. By removing these tumor escape mechanisms, sildenafil enhances intratumoral T cell infiltration and activation, reduces tumor outgrowth, and improves the antitumor efficacy of adoptive T cell therapy. Sildenafil also restores in vitro T cell proliferation of peripheral blood mononuclear cells from multiple myeloma and head and neck cancer patients. In light of the recent data that enzymes mediating MDSC-dependent immunosuppression in mice are active also in humans, these findings demonstrate a potentially novel use of PDE5 inhibitors as adjuncts to tumor-specific immune therapy.


Assuntos
3',5'-GMP Cíclico Fosfodiesterases/antagonistas & inibidores , Células Mieloides/citologia , Neoplasias/enzimologia , Neoplasias/prevenção & controle , Inibidores de Fosfodiesterase/farmacologia , Linfócitos T Reguladores/enzimologia , Linfócitos T Reguladores/imunologia , 3',5'-GMP Cíclico Fosfodiesterases/fisiologia , Animais , Antineoplásicos/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5 , Melanoma Experimental/enzimologia , Melanoma Experimental/imunologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Neoplasias/imunologia
8.
Immunol Rev ; 222: 162-79, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18364001

RESUMO

Emerging evidence indicates that the Achilles' heel of cancer immunotherapies is often the complex interplay of tumor-derived factors and deviant host properties, which involve a wide range of immune elements in the lymphoid and myeloid compartments. Regulatory lymphocytes, tumor-conditioned myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, and dysfunctional and immature dendritic cells take part in a complex immunoregulatory network. Despite the fact that some mechanisms governing tumor-induced immune tolerance and suppression are starting to be better understood and their complexity dissected, little is known about the diachronic picture of immune tolerance. Based on observations of MDSCs, we present a time-structured and topologically consistent idea of tumor-dependent tolerance progression in tumor-bearing hosts.


Assuntos
Tolerância Imunológica , Ativação Linfocitária , Células Mieloides/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Evasão Tumoral , Animais , Antígenos CD34/imunologia , Antígenos CD11/imunologia , Linfócitos T CD8-Positivos/imunologia , Comunicação Celular , Regulação para Baixo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Terapia de Imunossupressão , Subunidade alfa de Receptor de Interleucina-4/imunologia , Camundongos , Modelos Imunológicos , Mielopoese/imunologia , Neoplasias/patologia , Óxido Nítrico Sintase Tipo II/imunologia , Transdução de Sinais/imunologia
9.
J Immunother Cancer ; 10(1)2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35064009

RESUMO

BACKGROUND: Cancer-induced 'emergency' myelopoiesis plays a key role in tumor progression by inducing the accumulation of myeloid cells with a suppressive phenotype peripherally and in the tumor. Chemokine receptors (CCRs) and, in particular, CCR1, CCR2, CCR5, and CCR7 are emerging as key regulators of myeloid cell trafficking and function but their precise role has not been completely clarified yet because of the signal redundancy, integration, and promiscuity of chemokines and of the expression of these CCRs on other leukocyte subsets. METHODS: We used the 4PD nanoparticle for the in vivo targeted silencing of CCR1, CCR2, CCR5, and/or CCR7 in the myeloid cells of tumor bearing mice to evaluate the effect of treatments on tumor growth, myeloid cell trafficking and polarization. We used flow and image cytometry and functional assays to monitor changes in the tumor microenvironment and depletion experiments and immune deficient mice to determine the role of Ly6G+cells during tumor progression. We further evaluated in vitro the impact of chemokine receptor inhibition and tumor derived factors on myeloid cell differentiation from mouse and human hematopoietic stem and precursors cells (HSPCs) using flow cytometry, transcriptome analysis, cytokines beads arrays, functional assays, and mice deficient for CCR1 or CCR5. RESULTS: 4PD-mediated in vivo silencing of CCR1 and CCR5 on myeloid cells and myeloid precursors was necessary and sufficient to inhibit tumor progression. Functional studies indicated that this antitumor effect was not mediated by alteration of myeloid cell chemotaxes but rather by the repolarization of polymorphonuclear myeloid-derived suppressor cells (MDSCs) into tumoricidal neutrophils. Transcriptome functional and cytokine analysis indicated that tumor derived factors induced CCL3 and CCL4 in HSPCs that, through the autocrine engagement of CCR1 and CCR5, induced HSPCs differentiation in MDSCs. These finding were confirmed across mice with different genetic backgrounds and using HSPCs from umbilical cord blood and peripheral blood of patients with cancer. CONCLUSIONS: Our data support the notion that CCR1 and CCR5 and their ligands are a master immunological hub activated by several tumor derived factors. Activation of this pathway is necessary for the differentiation of MDSCs and protumoral macrophages.


Assuntos
Imunomodulação/imunologia , Células Supressoras Mieloides/metabolismo , Mielopoese/imunologia , Nanopartículas/metabolismo , Receptores CCR1/metabolismo , Receptores CCR5/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos , Microambiente Tumoral
10.
Nat Commun ; 13(1): 1815, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35383192

RESUMO

The ability to detect and target ß cells in vivo can substantially refine how diabetes is studied and treated. However, the lack of specific probes still hampers a precise characterization of human ß cell mass and the delivery of therapeutics in clinical settings. Here, we report the identification of two RNA aptamers that specifically and selectively recognize mouse and human ß cells. The putative targets of the two aptamers are transmembrane p24 trafficking protein 6 (TMED6) and clusterin (CLUS). When given systemically in immune deficient mice, these aptamers recognize the human islet graft producing a fluorescent signal proportional to the number of human islets transplanted. These aptamers cross-react with endogenous mouse ß cells and allow monitoring the rejection of mouse islet allografts. Finally, once conjugated to saRNA specific for X-linked inhibitor of apoptosis (XIAP), they can efficiently transfect non-dissociated human islets, prevent early graft loss, and improve the efficacy of human islet transplantation in immunodeficient in mice.


Assuntos
Aptâmeros de Nucleotídeos , Clusterina , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas , Proteínas de Transporte Vesicular , Animais , Aptâmeros de Nucleotídeos/genética , Clusterina/genética , Rejeição de Enxerto , Humanos , Indicadores e Reagentes , Ilhotas Pancreáticas/metabolismo , Camundongos , RNA/metabolismo , Proteínas de Transporte Vesicular/genética
11.
Cell Death Differ ; 29(2): 420-438, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34518653

RESUMO

Inflammatory responses rapidly detect pathogen invasion and mount a regulated reaction. However, dysregulated anti-pathogen immune responses can provoke life-threatening inflammatory pathologies collectively known as cytokine release syndrome (CRS), exemplified by key clinical phenotypes unearthed during the SARS-CoV-2 pandemic. The underlying pathophysiology of CRS remains elusive. We found that FLIP, a protein that controls caspase-8 death pathways, was highly expressed in myeloid cells of COVID-19 lungs. FLIP controlled CRS by fueling a STAT3-dependent inflammatory program. Indeed, constitutive expression of a viral FLIP homolog in myeloid cells triggered a STAT3-linked, progressive, and fatal inflammatory syndrome in mice, characterized by elevated cytokine output, lymphopenia, lung injury, and multiple organ dysfunctions that mimicked human CRS. As STAT3-targeting approaches relieved inflammation, immune disorders, and organ failures in these mice, targeted intervention towards this pathway could suppress the lethal CRS inflammatory state.


Assuntos
COVID-19/fisiopatologia , Síndrome da Liberação de Citocina/etiologia , Síndrome da Liberação de Citocina/metabolismo , Inflamação/metabolismo , Fator de Transcrição STAT3/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , COVID-19/metabolismo , Caspase 8/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , SARS-CoV-2/imunologia , Fator de Transcrição STAT3/genética , Transdução de Sinais
12.
Sci Transl Med ; 12(548)2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32554710

RESUMO

Local delivery of anticancer agents has the potential to maximize treatment efficacy and minimize the acute and long-term systemic toxicities. Here, we used unsupervised systematic evolution of ligands by exponential enrichment to identify four RNA aptamers that specifically recognized mouse and human myeloid cells infiltrating tumors but not their peripheral or circulating counterparts in multiple mouse models and from patients with head and neck squamous cell carcinoma (HNSCC). The use of these aptamers conjugated to doxorubicin enhanced the accumulation and bystander release of the chemotherapeutic drug in both primary and metastatic tumor sites in breast and fibrosarcoma mouse models. In the 4T1 mammary carcinoma model, these doxorubicin-conjugated aptamers outperformed Doxil, the first clinically approved highly optimized nanoparticle for targeted chemotherapy, promoting tumor regression after just three administrations with no detected changes in weight loss or blood chemistry. These RNA aptamers recognized tumor infiltrating myeloid cells in a variety of mouse tumors in vivo and from human HNSCC ex vivo. This work suggests the use of RNA aptamers for the detection of myeloid-derived suppressor cells in humans and for a targeted delivery of chemotherapy to the tumor microenvironment in multiple malignancies.


Assuntos
Antineoplásicos , Aptâmeros de Nucleotídeos , Neoplasias de Cabeça e Pescoço , Células Supressoras Mieloides , Animais , Linhagem Celular Tumoral , Humanos , Indicadores e Reagentes , Camundongos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Microambiente Tumoral
13.
J Clin Invest ; 116(10): 2777-90, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17016559

RESUMO

Active suppression of tumor-specific T lymphocytes can limit the efficacy of immune surveillance and immunotherapy. While tumor-recruited CD11b+ myeloid cells are known mediators of tumor-associated immune dysfunction, the true nature of these suppressive cells and the fine biochemical pathways governing their immunosuppressive activity remain elusive. Here we describe a population of circulating CD11b+IL-4 receptor alpha+ (CD11b+IL-4Ralpha+), inflammatory-type monocytes that is elicited by growing tumors and activated by IFN-gamma released from T lymphocytes. CD11b+IL-4Ralpha+ cells produced IL-13 and IFN-gamma and integrated the downstream signals of these cytokines to trigger the molecular pathways suppressing antigen-activated CD8+ T lymphocytes. Analogous immunosuppressive circuits were active in CD11b+ cells present within the tumor microenvironment. These suppressor cells challenge the current idea that tumor-conditioned immunosuppressive monocytes/macrophages are alternatively activated. Moreover, our data show how the inflammatory response elicited by tumors had detrimental effects on the adaptive immune system and suggest novel approaches for the treatment of tumor-induced immune dysfunctions.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Monócitos/imunologia , Neoplasias/imunologia , Animais , Arginase/genética , Arginase/metabolismo , Antígeno CD11b/análise , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Expressão Gênica/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imunidade Celular/imunologia , Interferon gama/genética , Interferon gama/metabolismo , Interferon gama/farmacologia , Interleucina-13/metabolismo , Interleucina-13/farmacologia , Interleucina-4/genética , Interleucina-4/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Imunológicos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Baço/citologia , Baço/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo
14.
Urology ; 132: 183-188, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31254572

RESUMO

OBJECTIVE: To present a configurable mathematical method to optimize long-term clinical decision-making for benign prostatic hyperplasia. METHODS: We designed a Markov chain model to simulate the different health states associated with benign prostatic hyperplasia and the transition between these states based on specific interventions: observation, pharmacotherapy, and 4 types of minimally invasive laser surgery. Transition probabilities, disutility scores, and costs for each health state were derived from the literature, expert opinion, and hospital administration data. Disutility was defined as the complement to one of the utility (1-utility), with utility representing the overall quality of life associated with a particular state. Linear programming was used to compute the Markov decision model. Primary outcomes include cost-effectiveness curves comparing the average treatment cost across permitted disutility levels while considering all modeled interventions. RESULTS: To achieve optimal patient outcomes (low International Prostate Symptoms Score), the model favored surgical interventions and increased costs of treatment. Between different desired disutility values (breakpoints), the model recommends performing 2 recommend treatments in relative proportions to achieve the lowest cost and optimal outcome. The model is limited by its theoretical basis and reliance on literature for transition probabilities and quality of life assessment. CONCLUSION: This model provides a tool for doctors, administrators, and patients to optimize cost-efficacy when considering multiple treatments and different severities of benign prostatic hyperplasia and may be configured to other disease states or clinical practices. Further studies are necessary to validate this model for real-life application.


Assuntos
Tomada de Decisão Clínica/métodos , Cadeias de Markov , Hiperplasia Prostática/terapia , Análise Custo-Benefício , Custos de Cuidados de Saúde , Humanos , Masculino , Modelos Estatísticos , Hiperplasia Prostática/economia
15.
Front Immunol ; 10: 1206, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31214178

RESUMO

Myeloid Derived suppressor cells (MDSCs) play a key role in the progression and recurrence of human malignancies and in restraining the efficacy of adjuvant therapies. We have previously shown that Tadalafil lowers MDSCs and regulatory T cells (Treg) in the blood and in the tumor, primes a tumor specific immune response, and increases the number of activated intratumoral CD8+T cells in patients with primary Head and Neck Squamous Cell Carcinoma (HNSCC). However, despite these important immune modulatory actions, to date no clinically significant effects have been reported following PDE5 inhibition. Here we report for the first time interim results of our ongoing phase I clinical trial (NCT02544880) in patients with recurrent HNSCC to evaluate the safety of and immunological effects of combining Tadalafil with the antitumor vaccine composed of Mucin1 (MUC1) and polyICLC. The combined treatment of Tadalafil and MUC1/polyICLC vaccine was well-tolerated with no serious adverse events or treatment limiting toxicities. Immunologically, this trial also confirms the positive immunomodulation of Tadalafil in patients with recurrent HNSCC and suggests an adjuvant effect of the anti-tumor vaccine MUC1/polyICLC. Additionally, image cytometry analysis of scanned tumors indicates that the PDE5 inhibitor Tadalafil in conjunction with the MUC1/polyICLC vaccine effectively reduces the number of PDL1+macrophages present at the tumor edge, and increases the number of activated tumor infiltrating T cells, suggesting reversion of immune exclusion. However, this analysis shows also that CD163 negative cells within the tumor upregulate PDL1 after treatment, suggesting the instauration of additional mechanisms of immune evasion. In summary, our data confirm the safety and immunologic potential of PDE5 inhibition in HNSCC but also point to PDL1 as additional mechanism of tumor evasion. This supports the rationale for combining checkpoint and PDE5 inhibitors for the treatment of human malignancies.


Assuntos
Antígeno B7-H1/genética , Vacinas Anticâncer/imunologia , Regulação Neoplásica da Expressão Gênica , Imunomodulação/efeitos dos fármacos , Carcinoma de Células Escamosas de Cabeça e Pescoço/etiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Tadalafila/administração & dosagem , Antígeno B7-H1/metabolismo , Biomarcadores , Vacinas Anticâncer/uso terapêutico , Terapia Combinada , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Estadiamento de Neoplasias , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Resultado do Tratamento
16.
Cancer Res ; 65(5): 2026-34, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15753403

RESUMO

A major limitation of adoptive immunotherapy is the availability of T cells specific for both terminally differentiated tumor cells and their clonogenic precursors. We show here that marrow-infiltrating lymphocytes (MILs) recognize myeloma cells after activation with anti-CD3/CD28 beads with higher frequency than activated peripheral blood lymphocytes from the same patients. Furthermore, activated MILs target both the terminally differentiated CD138+ plasma cells and the myeloma precursor as shown by profound inhibition in a tumor clonogenic assay. The presence of antigen in the marrow microenvironment seems to be important for the maintenance of tumor specificity. Taken together, these results highlight the intrinsic tumor specificity of MILs and describe a novel approach for the generation of tumor-specific T-cell populations suitable for adoptive immunotherapy of multiple myeloma.


Assuntos
Anticorpos Monoclonais/imunologia , Imunoterapia Adotiva , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Mieloma Múltiplo/terapia , Plasmócitos/imunologia , Adulto , Idoso , Anticorpos Monoclonais/uso terapêutico , Antígenos de Neoplasias/imunologia , Apoptose , Antígenos CD28/imunologia , Complexo CD3/imunologia , Caspases/metabolismo , Diferenciação Celular , Movimento Celular , Proliferação de Células , Feminino , Citometria de Fluxo , Humanos , Masculino , Glicoproteínas de Membrana/imunologia , Pessoa de Meia-Idade , Mieloma Múltiplo/imunologia , Proteoglicanas/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Sindecana-1 , Sindecanas , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
17.
Stud Health Technol Inform ; 123: 409-14, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17108460

RESUMO

Following total hip joint replacement (THJR), the durability of a prosthesis is limited by: wearing of frictional surfaces and loosening and migration of the prosthesis-cement-bone system. Literature review witnesses biomechanical studies focused mainly/only on hip functional state while none of them approached leg length discrepancy (LLD), posture unbalancing or spine related problems after THJR. Conversely, these latter could be critical elements for surgery and rehabilitation success, given the possible induction of asymmetric loading patterns. This study presents the results obtained by using a recently proposed methodology, to measure 3D subject posture balance and spine morphology and to evaluate its usefulness in individual therapy tuning/follow up. 3D subject's posture has been measured by means of 3D opto-electronic device, force platform and baropodography. 90 subjects after THJR have been included in this study. The subjects have been evaluated in two different epochs: 3 weeks after surgical intervention and after 3 months. 77/90 patients presented a LLD, pelvic obliquity and posture unbalancing. More than 90% of this group showed an overall postural re-balancing induced by the use of simple underfoot wedge. 70/77 patients needed wedge under the healthy side showing the surgical intervention produced a leg lengthening. 60/90 (52 LLD) patients underwent up to now to control and patients who wore the suggested wedge (63.4%) presented an improvement over all the considered quantitative parameters. Patients who wore a shorter than suggested wedge (23.1%), or that did not wear the suggested wedge (13.5%) presented a moderate or significant worsening of their postural balancing respectively.


Assuntos
Artroplastia de Quadril , Imageamento Tridimensional , Desigualdade de Membros Inferiores , Suporte de Carga , Adulto , Idoso , Fenômenos Biomecânicos , Humanos , Pessoa de Meia-Idade , Postura , Próteses e Implantes
18.
Cancer Res ; 64(17): 6337-43, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15342423

RESUMO

Tumor vaccines have shown promise in early clinical trials. Among them, tumor cells genetically engineered to secrete biologically active granulocyte-macrophage colony-stimulating factor (GM-CSF) can generate a systemic antitumor immune response. Although the minimal required GM-CSF dose produced by modified tumor cells to achieve a measurable antitumor effect is well known, no data examined whether an upper therapeutic limit may exist for this vaccination strategy. Because recent data demonstrate an immunosuppressive effect of GM-CSF produced by growing tumors, we thus sought to determine whether high GM-CSF doses administered in a vaccine formulation could impair antitumor immunity. Using a vaccine strategy involving a GM-CSF-producing bystander cell line (B78H1-GM) admixed with autologous tumor, we assessed the impact of varying doses of GM-CSF while maintaining a constant antigen dose. Our results defined a threshold above which a GM-CSF-based vaccine not only lost its efficacy, but more importantly for its clinical implications resulted in substantial immunosuppression in vivo. Above this threshold, GM-CSF induced Gr1+/CD11b+ myeloid suppressor cells that substantially impaired antigen-specific T-cell responses and adversely affected antitumor immune responses in vivo. The dual effects of GM-CSF are mediated by the systemic and not local concentration of this cytokine. Myeloid suppressor cell-induced immunosuppression is mediated by nitric oxide production via inducible nitric oxide synthase (iNOS) because the specific iNOS inhibitor, l-NMMA, restored antigen-specific T-cell responsiveness in vitro. Taken together, our data demonstrated the negative impact of supra-therapeutic vaccine doses of GM-CSF and underscored the importance of identifying these critical variables in an effort to increase the therapeutic efficacy of tumor vaccines.


Assuntos
Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Células Mieloides/imunologia , Neoplasias Experimentais/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Relação Dose-Resposta Imunológica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Neoplasias Experimentais/terapia
19.
Cancer Res ; 64(21): 8068-76, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15520218

RESUMO

Definition of immune variables that correlate with the antitumor activity of cancer vaccines is critical for monitoring immunotherapy protocols. To define surrogate end points predictive of the therapeutic efficacy of recombinant vaccines based on melanoma antigen tyrosinase-related protein (TRP)-2, we evaluated several properties of antigen-specific CD8(+) T lymphocytes in single mice undergoing either prophylactic or therapeutic immunization. Predictive markers for the efficacy of genetic vaccination were identified in the prophylactic model used. Interestingly, the number of tetramer(+) CD8(+) T lymphocytes expanded in vitro after a single cycle of stimulation with the immunodominant TRP-2 peptide was of the highest predictive value. In the therapeutic model, no variable examined at a single mouse level predicted the long-term therapeutic effect. Mice that survived did not show the highest expansion of antigen-specific lymphocytes or the more functionally active effectors, ex vivo or after in vitro culture with the peptide antigen. Successful therapy correlated strictly with the skewing of the T-cell receptor repertoire of tetramer-sorted, TRP-2-specific CD8(+) T lymphocytes, which showed a preferential alpha chain usage with a common CDR3 region.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Rearranjo Gênico da Cadeia alfa dos Receptores de Antígenos dos Linfócitos T , Melanoma/imunologia , Vacinas Sintéticas/uso terapêutico , Adenoviridae/genética , Animais , Imunização , Oxirredutases Intramoleculares/imunologia , Camundongos , Camundongos Endogâmicos C57BL
20.
Vaccines (Basel) ; 4(3)2016 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-27618112

RESUMO

Neutrophils are the most abundant circulating blood cell type in humans, and are the first white blood cells recruited at the inflammation site where they orchestrate the initial immune response. Although their presence at the tumor site was recognized in the 1970s, until recently these cells have been neglected and considered to play just a neutral role in tumor progression. Indeed, in recent years neutrophils have been recognized to play a dual role in tumor development by either assisting the growth, angiogenesis, invasion, and metastasis or by exerting tumoricidal action directly via the secretion of antitumoral compounds, or indirectly via the orchestration of antitumor immunity. Understanding the biology of these cells and influencing their polarization in the tumor micro- and macro-environment may be the key for the development of new therapeutic strategies, which may finally hold the promise of an effective immunotherapy for cancer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA