Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
N Engl J Med ; 387(23): 2113-2125, 2022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36477031

RESUMO

BACKGROUND: Immune checkpoint inhibitors and targeted therapies have dramatically improved outcomes in patients with advanced melanoma, but approximately half these patients will not have a durable benefit. Phase 1-2 trials of adoptive cell therapy with tumor-infiltrating lymphocytes (TILs) have shown promising responses, but data from phase 3 trials are lacking to determine the role of TILs in treating advanced melanoma. METHODS: In this phase 3, multicenter, open-label trial, we randomly assigned patients with unresectable stage IIIC or IV melanoma in a 1:1 ratio to receive TIL or anti-cytotoxic T-lymphocyte antigen 4 therapy (ipilimumab at 3 mg per kilogram of body weight). Infusion of at least 5×109 TILs was preceded by nonmyeloablative, lymphodepleting chemotherapy (cyclophosphamide plus fludarabine) and followed by high-dose interleukin-2. The primary end point was progression-free survival. RESULTS: A total of 168 patients (86% with disease refractory to anti-programmed death 1 treatment) were assigned to receive TILs (84 patients) or ipilimumab (84 patients). In the intention-to-treat population, median progression-free survival was 7.2 months (95% confidence interval [CI], 4.2 to 13.1) in the TIL group and 3.1 months (95% CI, 3.0 to 4.3) in the ipilimumab group (hazard ratio for progression or death, 0.50; 95% CI, 0.35 to 0.72; P<0.001); 49% (95% CI, 38 to 60) and 21% (95% CI, 13 to 32) of the patients, respectively, had an objective response. Median overall survival was 25.8 months (95% CI, 18.2 to not reached) in the TIL group and 18.9 months (95% CI, 13.8 to 32.6) in the ipilimumab group. Treatment-related adverse events of grade 3 or higher occurred in all patients who received TILs and in 57% of those who received ipilimumab; in the TIL group, these events were mainly chemotherapy-related myelosuppression. CONCLUSIONS: In patients with advanced melanoma, progression-free survival was significantly longer among those who received TIL therapy than among those who received ipilimumab. (Funded by the Dutch Cancer Society and others; ClinicalTrials.gov number, NCT02278887.).


Assuntos
Imunoterapia Adotiva , Linfócitos do Interstício Tumoral , Melanoma , Humanos , Terapia Baseada em Transplante de Células e Tecidos , Ipilimumab/efeitos adversos , Melanoma/tratamento farmacológico
2.
Cell Microbiol ; 15(12): 2093-108, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23902464

RESUMO

The causative agent of tuberculosis (TB), Mycobacterium tuberculosis, remains an important worldwide health threat. Although TB is one of the oldest infectious diseases of man, a detailed understanding of the mycobacterial mechanisms underlying pathogenesis remains elusive. Here, we studied the role of the α(1→2) mannosyltransferase MptC in mycobacterial virulence, using the Mycobacterium marinum zebrafish infection model. Like its M. tuberculosis orthologue, disruption of M. marinum mptC (mmar_3225) results in defective elongation of mannose caps of lipoarabinomannan (LAM) and absence of α(1→2)mannose branches on the lipomannan (LM) and LAM mannan core, as determined by biochemical analysis (NMR and GC-MS) and immunoblotting. We found that the M. marinum mptC mutant is strongly attenuated in embryonic zebrafish, which rely solely on innate immunity, whereas minor virulence defects were observed in adult zebrafish. Strikingly, complementation with the Mycobacterium smegmatis mptC orthologue, which restored mannan core branching but not cap elongation, was sufficient to fully complement the virulence defect of the mptC mutant in embryos. Altogether our data demonstrate that not LAM capping, but mannan core branching of LM/LAM plays an important role in mycobacterial pathogenesis in the context of innate immunity.


Assuntos
Lipopolissacarídeos/metabolismo , Mycobacterium marinum/imunologia , Mycobacterium marinum/patogenicidade , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/metabolismo , Animais , Carga Bacteriana , Imunidade Inata , Lipopolissacarídeos/química , Manose/química , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium marinum/genética , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/genética , Tuberculose/imunologia , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia
3.
Cell Microbiol ; 14(8): 1287-98, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22524898

RESUMO

Mycobacterium species, including Mycobacterium tuberculosis and Mycobacterium leprae, are among the most potent human bacterial pathogens. The discovery of cytosolic mycobacteria challenged the paradigm that these pathogens exclusively localize within the phagosome of host cells. As yet the biological relevance of mycobacterial translocation to the cytosol remained unclear. In this current study we used electron microscopy techniques to establish a clear link between translocation and mycobacterial virulence. Pathogenic, patient-derived mycobacteria species were found to translocate to the cytosol, while non-pathogenic species did not. We were further able to link cytosolic translocation with pathogenicity by introducing the ESX-1 (type VII) secretion system into the non-virulent, exclusively phagolysosomal Mycobacterium bovis BCG. Furthermore, we show that translocation is dependent on the C-terminus of the early-secreted antigen ESAT-6. The C-terminal truncation of ESAT-6 was shown to result in attenuation in mice, again linking translocation to virulence. Together, these data demonstrate the molecular mechanism facilitating translocation of mycobacteria. The ability to translocate from the phagolysosome to the cytosol is with this study proven to be biologically significant as it determines mycobacterial virulence.


Assuntos
Citoplasma/microbiologia , Mycobacterium/patogenicidade , Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Linhagem Celular , Técnicas de Introdução de Genes , Interações Hospedeiro-Patógeno , Humanos , Lisossomos/microbiologia , Lisossomos/ultraestrutura , Mycobacterium/genética , Mycobacterium/metabolismo , Fagossomos/microbiologia , Fagossomos/ultraestrutura , Estrutura Terciária de Proteína , Ubiquitina/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
4.
J Immunol ; 187(9): 4744-53, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21957139

RESUMO

During infection of humans and animals, pathogenic mycobacteria manipulate the host cell causing severe diseases such as tuberculosis and leprosy. To understand the basis of mycobacterial pathogenicity, it is crucial to identify the molecular virulence mechanisms. In this study, we address the contribution of ESX-1 and ESX-5--two homologous type VII secretion systems of mycobacteria that secrete distinct sets of immune modulators--during the macrophage infection cycle. Using wild-type, ESX-1- and ESX-5-deficient mycobacterial strains, we demonstrate that these secretion systems differentially affect subcellular localization and macrophage cell responses. We show that in contrast to ESX-1, the effector proteins secreted by ESX-5 are not required for the translocation of Mycobacterium tuberculosis or Mycobacterium marinum to the cytosol of host cells. However, the M. marinum ESX-5 mutant does not induce inflammasome activation and IL-1ß activation. The ESX-5 system also induces a caspase-independent cell death after translocation has taken place. Importantly, by means of inhibitory agents and small interfering RNA experiments, we reveal that cathepsin B is involved in both the induction of cell death and inflammasome activation upon infection with wild-type mycobacteria. These results reveal distinct roles for two different type VII secretion systems during infection and shed light on how virulent mycobacteria manipulate the host cell in various ways to replicate and spread.


Assuntos
Proteínas de Homeodomínio/metabolismo , Inflamassomos/imunologia , Inflamassomos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Mycobacterium marinum/imunologia , Mycobacterium tuberculosis/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Morte Celular/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Interleucina-1beta/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , Camundongos , Mycobacterium marinum/patogenicidade , Mycobacterium tuberculosis/patogenicidade
5.
PLoS Pathog ; 6(3): e1000794, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20221442

RESUMO

The cell envelope of mycobacteria, a group of Gram positive bacteria, is composed of a plasma membrane and a Gram-negative-like outer membrane containing mycolic acids. In addition, the surface of the mycobacteria is coated with an ill-characterized layer of extractable, non-covalently linked glycans, lipids and proteins, collectively known as the capsule, whose occurrence is a matter of debate. By using plunge freezing cryo-electron microscopy technique, we were able to show that pathogenic mycobacteria produce a thick capsule, only present when the cells were grown under unperturbed conditions and easily removed by mild detergents. This detergent-labile capsule layer contains arabinomannan, alpha-glucan and oligomannosyl-capped glycolipids. Further immunogenic and proteomic analyses revealed that Mycobacterium marinum capsule contains high amounts of proteins that are secreted via the ESX-1 pathway. Finally, cell infection experiments demonstrated the importance of the capsule for binding to cells and dampening of pro-inflammatory cytokine response. Together, these results show a direct visualization of the mycobacterial capsular layer as a labile structure that contains ESX-1-secreted proteins.


Assuntos
Cápsulas Bacterianas/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Macrófagos/microbiologia , Mycobacterium/metabolismo , Mycobacterium/ultraestrutura , Cápsulas Bacterianas/ultraestrutura , Proteínas da Membrana Bacteriana Externa/ultraestrutura , Microscopia Crioeletrônica , Citocinas/metabolismo , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Microscopia Imunoeletrônica , Mycobacterium bovis/metabolismo , Mycobacterium bovis/ultraestrutura , Mycobacterium marinum/metabolismo , Mycobacterium marinum/ultraestrutura , Mycobacterium smegmatis/metabolismo , Mycobacterium smegmatis/ultraestrutura , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/ultraestrutura
6.
Proc Natl Acad Sci U S A ; 105(32): 11281-6, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18685099

RESUMO

Dendritic cells (DC) present lipid and peptide antigens to T cells on CD1 and MHC Class II (MHCII), respectively. The relative contribution of these systems during the initiation of adaptive immunity after microbial infection is not characterized. MHCII molecules normally acquire antigen and rapidly traffic from phagolysosomes to the plasma membrane as part of DC maturation, whereas CD1 molecules instead continually recycle between these sites before, during, and after DC maturation. We find that in Mycobacterium tuberculosis (Mtb)-infected DCs, CD1 presents antigens quickly. Surprisingly, rapid DC maturation results in early failure and delay in MHCII presentation. Whereas both CD1b and MHCII localize to bacterial phagosomes early after phagocytosis, MHCII traffics from the phagosome to the plasma membrane with a rapid kinetic that can precede antigen availability and loading. Thus, rather than facilitating antigen presentation, a lack of coordination in timing may allow organisms to use DC maturation as a mechanism of immune evasion. In contrast, CD1 antigen presentation occurs in the face of Mtb infection and rapid DC maturation because a pool of CD1 molecules remains available on the phagolysosome membrane that is able to acquire lipid antigens and deliver them to the plasma membrane.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos CD1/imunologia , Células Dendríticas/imunologia , Lipídeos/imunologia , Mycobacterium tuberculosis/imunologia , Peptídeos/imunologia , Tuberculose/imunologia , Transporte Biológico Ativo/imunologia , Membrana Celular/imunologia , Células Cultivadas , Células Dendríticas/microbiologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Cinética , Fagocitose/imunologia , Fagossomos/imunologia , Linfócitos T/imunologia
7.
Cell Rep ; 37(7): 110013, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34788605

RESUMO

Autotaxin (ATX; ENPP2) produces lysophosphatidic acid (LPA) that regulates multiple biological functions via cognate G protein-coupled receptors LPAR1-6. ATX/LPA promotes tumor cell migration and metastasis via LPAR1 and T cell motility via LPAR2, yet its actions in the tumor immune microenvironment remain unclear. Here, we show that ATX secreted by melanoma cells is chemorepulsive for tumor-infiltrating lymphocytes (TILs) and circulating CD8+ T cells ex vivo, with ATX functioning as an LPA-producing chaperone. Mechanistically, T cell repulsion predominantly involves Gα12/13-coupled LPAR6. Upon anti-cancer vaccination of tumor-bearing mice, ATX does not affect the induction of systemic T cell responses but, importantly, suppresses tumor infiltration of cytotoxic CD8+ T cells and thereby impairs tumor regression. Moreover, single-cell data from melanoma tumors are consistent with intratumoral ATX acting as a T cell repellent. These findings highlight an unexpected role for the pro-metastatic ATX-LPAR axis in suppressing CD8+ T cell infiltration to impede anti-tumor immunity, suggesting new therapeutic opportunities.


Assuntos
Linfócitos do Interstício Tumoral/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Quimiotaxia/fisiologia , Feminino , Humanos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias , Diester Fosfórico Hidrolases/fisiologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/fisiologia , Microambiente Tumoral
8.
mSphere ; 6(3)2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33952660

RESUMO

Mycobacterium tuberculosis infections claim more than a million lives each year, and better treatments or vaccines are required. A crucial pathogenicity factor is translocation from phagolysosomes to the cytosol upon phagocytosis by macrophages. Translocation from the phagolysosome to the cytosol is an ESX-1-dependent process, as previously shown in vitro Here, we show that in vivo, mycobacteria also translocate to the cytosol but mainly when host immunity is compromised. We observed only low numbers of cytosolic bacilli in mice, armadillos, zebrafish, and patient material infected with M. tuberculosis, M. marinum, or M. leprae In contrast, when innate or adaptive immunity was compromised, as in severe combined immunodeficiency (SCID) or interleukin-1 receptor 1 (IL-1R1)-deficient mice, significant numbers of cytosolic M. tuberculosis bacilli were detected in the lungs of infected mice. Taken together, in vivo, translocation to the cytosol of M. tuberculosis is controlled by adaptive immune responses as well as IL-1R1-mediated signals.IMPORTANCE For decades, Mycobacterium tuberculosis has been one of the deadliest pathogens known. Despite infecting approximately one-third of the human population, no effective treatment or vaccine is available. A crucial pathogenicity factor is subcellular localization, as M. tuberculosis can translocate from phagolysosome to the cytosol in macrophages. The situation in vivo is more complicated. In this study, we establish that high-level cytosolic escape of mycobacteria can indeed occur in vivo but mainly when host resistance is compromised. The IL-1 pathway is crucial for the control of the number of cytosolic mycobacteria. The establishment that immune signals result in the clearance of cells containing cytosolic mycobacteria connects two important fields, cell biology and immunology, which is vital for the understanding of the pathology of M. tuberculosis.


Assuntos
Citosol/microbiologia , Mycobacterium/imunologia , Mycobacterium/patogenicidade , Fagossomos/microbiologia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Transdução de Sinais/imunologia , Animais , Tatus/microbiologia , Translocação Bacteriana , Citosol/imunologia , Feminino , Humanos , Hanseníase/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Mycobacterium/classificação , Fagossomos/imunologia , Pele/microbiologia , Pele/patologia , Células THP-1 , Peixe-Zebra
9.
J Immunol ; 181(10): 7166-75, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18981138

RESUMO

The ESX-5 secretion system of pathogenic mycobacteria is responsible for the secretion of various PPE and PE-PGRS proteins. To better understand the role of ESX-5 effector proteins in virulence, we analyzed the interactions of Mycobacterium marinum ESX-5 mutant with human macrophages (Mphi). Both wild-type bacteria and the ESX-5 mutant were internalized and the ESX-5 mutation did not affect the escape of mycobacteria from phagolysosomes into the cytosol, as was shown by electron microscopy. However, the ESX-5 mutation strongly effected expression of surface Ags and cytokine secretion. Whereas wild-type M. marinum actively suppressed the induction of appreciable levels of IL-12p40, TNF-alpha, and IL-6, infection with the ESX-5 mutant resulted in strongly induced production of these proinflammatory cytokines. By contrast, infection with M. marinum wild-type strain resulted in a significant induction of IL-1beta production as compared with the ESX-5 mutant. These results show that ESX-5 plays an essential role in the modulation of immune cytokine secretion by human Mphi. Subsequently, we show that an intact ESX-5 secretion system actively suppresses TLR signaling-dependent innate immune cytokine secretion. Together, our results show that ESX-5 substrates, directly or indirectly, strongly modulate the human Mphi response at various critical steps.


Assuntos
Proteínas de Bactérias/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium marinum/patogenicidade , Fatores de Virulência/imunologia , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/imunologia , Células Cultivadas , Citocinas/biossíntese , Humanos , Immunoblotting , Fatores de Virulência/metabolismo
10.
J Immunother Cancer ; 8(2)2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32753545

RESUMO

Treatment of metastatic melanoma with autologous tumor infiltrating lymphocytes (TILs) is currently applied in several centers. Robust and remarkably consistent overall response rates, of around 50% of treated patients, have been observed across hospitals, including a substantial fraction of durable, complete responses. PURPOSE: Execute a phase I/II feasibility study with TIL therapy in metastatic melanoma at the Netherlands Cancer Institute, with the goal to assess feasibility and potential value of a randomized phase III trial. EXPERIMENTAL: Ten patients were treated with TIL therapy. Infusion products and peripheral blood samples were phenotypically characterized and neoantigen reactivity was assessed. Here, we present long-term clinical outcome and translational data on neoantigen reactivity of the T cell products. RESULTS: Five out of 10 patients, who were all anti-PD-1 naïve at time of treatment, showed an objective clinical response, including two patients with a complete response that are both ongoing for more than 7 years. Immune monitoring demonstrated that neoantigen-specific T cells were detectable in TIL infusion products from three out of three patients analyzed. For six out of the nine neoantigen-specific T cell responses detected in these TIL products, T cell response magnitude increased significantly in the peripheral blood compartment after therapy, and neoantigen-specific T cells were detectable for up to 3 years after TIL infusion. CONCLUSION: The clinical results from this study confirm the robustness of TIL therapy in metastatic melanoma and the potential role of neoantigen-specific T cell reactivity. In addition, the data from this study supported the rationale to initiate an ongoing multicenter phase III TIL trial.


Assuntos
Linfócitos do Interstício Tumoral/metabolismo , Melanoma/genética , Linfócitos T/metabolismo , Adulto , Idoso , Feminino , Seguimentos , Humanos , Masculino , Melanoma/patologia , Pessoa de Meia-Idade
11.
Cell ; 129(7): 1287-98, 2007 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-17604718

RESUMO

M. tuberculosis and M. leprae are considered to be prototypical intracellular pathogens that have evolved strategies to enable growth in the intracellular phagosomes. In contrast, we show that lysosomes rapidly fuse with the virulent M. tuberculosis- and M. leprae-containing phagosomes of human monocyte-derived dendritic cells and macrophages. After 2 days, M. tuberculosis progressively translocates from phagolysosomes into the cytosol in nonapoptotic cells. Cytosolic entry is also observed for M. leprae but not for vaccine strains such as M. bovis BCG or in heat-killed mycobacteria and is dependent upon secretion of the mycobacterial gene products CFP-10 and ESAT-6. The cytosolic bacterial localization and replication are pathogenic features of virulent mycobacteria, causing significant cell death within a week. This may also reveal a mechanism for MHC-based antigen presentation that is lacking in current vaccine strains.


Assuntos
Citosol/fisiologia , Lisossomos/fisiologia , Mycobacterium/fisiologia , Células Mieloides/microbiologia , Fagossomos/fisiologia , Apresentação de Antígeno/fisiologia , Biomarcadores/metabolismo , Compartimento Celular/fisiologia , Morte Celular/fisiologia , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Citosol/ultraestrutura , Regulação Bacteriana da Expressão Gênica/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Humanos , Imuno-Histoquímica , Membranas Intracelulares/fisiologia , Membranas Intracelulares/ultraestrutura , Lisossomos/ultraestrutura , Proteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Mycobacterium/genética , Mycobacterium/ultraestrutura , Mycobacterium leprae/genética , Mycobacterium leprae/fisiologia , Mycobacterium leprae/ultraestrutura , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/fisiologia , Mycobacterium tuberculosis/ultraestrutura , Células Mieloides/fisiologia , Células Mieloides/ultraestrutura , Fagossomos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA