Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(40): 20141-20150, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31527243

RESUMO

Tumors utilize a number of effective strategies, including the programmed death 1/PD ligand 1 (PD-1/PD-L1) axis, to evade immune-mediated control of their growth. PD-L1 expression is mainly induced by IFN receptor signaling or constitutively induced. Integrins are an abundantly expressed class of proteins which play multiple deleterious roles in cancer and exert proangiogenic and prosurvival activities. We asked whether αvß3-integrin positively regulates PD-L1 expression and the anticancer immune response. We report that αvß3-integrin regulated constitutive and IFN-induced PD-L1 expression in human and murine cancerous and noncancerous cells. αvß3-integrin targeted STAT1 through its signaling C tail. The implantation of ß3-integrin-depleted tumor cells led to a dramatic decrease in the growth of primary tumors, which exhibited reduced PD-L1 expression and became immunologically hot, with increased IFNγ content and CD8+ cell infiltration. In addition, the implantation of ß3-integrin-depleted tumors elicited an abscopal immunotherapeutic effect measured as protection from the challenge tumor and durable splenocyte and serum reactivity to B16 cell antigens. These modifications to the immunosuppressive microenvironment primed cells for checkpoint (CP) blockade. When combined with anti-PD-1, ß3-integrin depletion led to durable therapy and elicited an abscopal immunotherapeutic effect. We conclude that in addition to its previously known roles, αvß3-integrin serves as a critical component of the cancer immune evasion strategy and can be an effective immunotherapy target.

2.
Adv Exp Med Biol ; 1290: 67-80, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33559855

RESUMO

The great hopes raised by the discovery of the immunoregulatory cytokine interleukin 12 (IL-12) as an anticancer agent were marred during early clinical experimentation because of severe adverse effects, which prompted a search for alternative formulations and routes of administration. Onco-immunotherapeutic viruses (OIVs) are wild-type or genetically engineered viruses that exert antitumor activity by causing death of the tumor cells they infect and by overcoming a variety of immunosuppressive mechanisms put in place by the tumors. OIVs have renewed the interest in IL-12, as they offer the opportunity to encode the cytokine transgenically from the viral genome and to produce it at high concentrations in the tumor bed. A large body of evidence indicates that IL-12 serves as a potent adjuvant for the immunotherapeutic response elicited by OIVs in murine tumor models. The list of OIVs includes onco-immunotherapeutic herpes simplex, adeno, measles, Newcastle disease, and Maraba viruses, among others. The large increase in IL-12-mediated adjuvanticity was invariably observed for all the OIVs analyzed. Indirect evidence suggests that locally delivered IL-12 may also increase tumor antigenicity. Importantly, the OIV/IL-12 treatment was not accompanied by adverse effects and elicited a long-lasting immune response capable of halting the growth of distant tumors. Thus, OIVs provide an avenue for reducing the clinical toxicity associated with systemic IL-12 therapy, by concentrating the cytokine at the site of disease. The changes to the tumor microenvironment induced by the IL-12-armed OIVs primed the tumors to an improved response to the checkpoint blockade therapy, suggesting that the triple combination is worth pursuing in the future. The highly encouraging results in preclinical models have prompted translation to the clinic. How well the IL-12-OIV-checkpoint inhibitors' combination will perform in humans remains to be fully investigated.


Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Humanos , Imunoterapia , Interleucina-12/genética , Camundongos , Vírus Oncolíticos/genética , Microambiente Tumoral
3.
PLoS Pathog ; 14(8): e1007209, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30080893

RESUMO

Oncolytic herpes simplex viruses (oHSVs) showed efficacy in clinical trials and practice. Most of them gain cancer-specificity from deletions/mutations in genes that counteract the host response, and grow selectively in cancer cells defective in anti-viral response. Because of the deletions/mutations, they are frequently attenuated or over-attenuated. We developed next-generation oHSVs, which carry no deletion/mutation, gain cancer-specificity from specific retargeting to tumor cell receptors-e.g. HER2 (human epidermal growth factor receptor 2)-hence are fully-virulent in the targeted cancer cells. The type of immunotherapy they elicit was not predictable, since non-attenuated HSVs induce and then dampen the innate response, whereas deleted/attenuated viruses fail to contrast it, and since the retargeted oHSVs replicate efficiently in tumor cells, but spare other cells in the tumor. We report on the first efficacy study of HER2-retargeted, fully-virulent oHSVs in immunocompetent mice. Their safety profile was very high. Both the unarmed R-LM113 and the IL-12-armed R-115 inhibited the growth of the primary HER2-Lewis lung carcinoma-1 (HER2-LLC1) tumor, R-115 being constantly more efficacious. All the mice that did not die because of the primary treated tumors, were protected from the growth of contralateral untreated tumors. The long-term survivors were protected from a second contralateral tumor, providing additional evidence for an abscopal immunotherapeutic effect. Analysis of the local response highlighted that particularly R-115 unleashed the immunosuppressive tumor microenvironment, i.e. induced immunomodulatory cytokines, including IFNγ, T-bet which promoted Th1 polarization. Some of the tumor infiltrating cells, e.g. CD4+, CD335+ cells were increased in the tumors of all responders mice, irrespective of which virus was employed, whereas CD8+, Foxp3+, CD141+ were increased and CD11b+ cells were decreased preferentially in R-115-treated mice. The durable response included a breakage of tolerance towards both HER2 and the wt tumor cells, and underscored a systemic immunotherapeutic vaccine response.


Assuntos
Antineoplásicos/farmacologia , Vacinas Anticâncer/farmacologia , Imunoterapia Ativa/métodos , Interleucina-12 , Terapia Viral Oncolítica/métodos , Simplexvirus , Animais , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Camundongos , Vírus Oncolíticos
4.
J Virol ; 92(6)2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29263255

RESUMO

Insertion of a single-chain variable-fragment antibody (scFv) to HER2 (human epidermal growth factor receptor 2) in gD, gH, or gB gives rise to herpes simplex viruses (HSVs) specifically retargeted to HER2-positive cancer cells, hence to highly specific nonattenuated oncolytic agents. Clinical-grade virus production cannot rely on cancer cells. Recently, we developed a double-retargeting strategy whereby gH carries the GCN4 peptide for retargeting to the noncancer producer Vero-GCN4R cell line and gD carries the scFv to HER2 for cancer retargeting. Here, we engineered double-retargeted recombinants, which carry both the GCN4 peptide and the scFv to HER2 in gD. Novel, more-advantageous detargeting strategies were devised so as to optimize the cultivation of the double-retargeted recombinants. Nectin1 detargeting was achieved by deletion of amino acids (aa) 35 to 39, 214 to 223, or 219 to 223 and replacement of the deleted sequences with one of the two ligands. The last two deletions were not attempted before. All recombinants exhibited the double retargeting to HER2 and to the Vero-GCN4R cells, as well as detargeting from the natural receptors HVEM and nectin1. Of note, some recombinants grew to higher yields than others. The best-performing recombinants carried a gD deletion as small as 5 amino acids and grew to titers similar to those exhibited by the singly retargeted R-LM113 and by the nonretargeted R-LM5. This study shows that double retargeting through insertion of two ligands in gD is feasible and, when combined with appropriate detargeting modifications, can result in recombinants highly effective in vitro and in vivoIMPORTANCE There is increasing interest in oncolytic viruses following the FDA and European Medicines Agency (EMA) approval of the oncolytic HSV OncovexGM-CSF and, mainly, because they greatly boost the immune response to the tumor and can be combined with immunotherapeutic agents, particularly immune checkpoint inhibitors. A strategy to gain high cancer specificity and avoid virus attenuation is to retarget the virus tropism to cancer-specific receptors of choice. However, cultivation of retargeted oncolytics in cells expressing the cancer receptor may not be approvable by regulatory agencies. We devised a strategy for their cultivation in noncancer cells. Here, we describe a double-retargeting strategy, based on the simultaneous insertion of two ligands in gD, one for retargeting to a producer, universal Vero cell derivative and one for retargeting to the HER2 cancer receptor. These insertions were combined with novel, minimally disadvantageous detargeting modifications. The current and accompanying studies indicate how to best achieve the clinical-grade cultivation of retargeted oncolytics.


Assuntos
Vírus Oncolíticos , Peptídeos , Proteínas do Envelope Viral , Tropismo Viral/genética , Animais , Chlorocebus aethiops , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
5.
J Virol ; 92(6)2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29263257

RESUMO

Oncolytic viruses gain cancer specificity in several ways. Like the majority of viruses, they grow better in cancer cells that are defective in mounting the host response to viruses. Often, they are attenuated by deletion or mutation of virulence genes that counteract the host response or are naturally occurring oncolytic mutants. In contrast, retargeted viruses are not attenuated or deleted; their cancer specificity rests on a modified, specific tropism for cancer receptors. For herpes simplex virus (HSV)-based oncolytics, the detargeting-retargeting strategies employed so far were based on genetic modifications of gD. Recently, we showed that even gH or gB can serve as retargeting tools. To enable the growth of retargeted HSVs in cells that can be used for clinical-grade virus production, a double-retargeting strategy has been developed. Here we show that several sites in the N terminus of gB are suitable to harbor the 20-amino-acid (aa)-long GCN4 peptide, which readdresses HSV tropism to Vero cells expressing the artificial GCN4 receptor and thus enables virus cultivation in the producer noncancer Vero-GCN4R cell line. The gB modifications can be combined with a minimal detargeting modification in gD, consisting in the deletion of two residues, aa 30 and 38, and replacement of aa 38 with the scFv to human epidermal growth factor receptor 2 (HER2), for retargeting to the cancer receptor. The panel of recombinants was analyzed comparatively in terms of virus growth, cell-to-cell spread, cytotoxicity, and in vivo antitumor efficacy to define the best double-retargeting strategy.IMPORTANCE There is increasing interest in oncolytic viruses, following FDA and the European Medicines Agency (EMA) approval of HSV OncovexGM-CSF, and, mainly, because they greatly boost the immune response to the tumor and can be combined with immunotherapeutic agents, particularly checkpoint inhibitors. A strategy to gain cancer specificity and avoid virus attenuation is to retarget the virus tropism to cancer-specific receptors of choice. Cultivation of fully retargeted viruses is challenging, since they require cells that express the cancer receptor. We devised a strategy for their cultivation in producer noncancer Vero cell derivatives. Here, we developed a double-retargeting strategy, based on insertion of one ligand in gB for retargeting to a Vero cell derivative and of anti-HER2 ligand in gD for cancer retargeting. These modifications were combined with a minimally destructive detargeting strategy. This study and its companion paper explain the clinical-grade cultivation of retargeted oncolytic HSVs and promote their translation to the clinic.


Assuntos
Herpesvirus Humano 1 , Neoplasias Experimentais/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Anticorpos de Cadeia Única , Proteínas do Envelope Viral , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
PLoS Pathog ; 13(4): e1006352, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28423057

RESUMO

Herpes simplex virus (HSV) entry into the cells requires glycoproteins gD, gH/gL and gB, activated in a cascade fashion by conformational modifications induced by cognate receptors and intermolecular signaling. The receptors are nectin1 and HVEM (Herpes virus entry mediator) for gD, and αvß6 or αvß8 integrin for gH. In earlier work, insertion of a single chain antibody (scFv) to the cancer receptor HER2 (human epidermal growth factor receptor 2) in gD, or in gH, resulted in HSVs specifically retargeted to the HER2-positive cancer cells, hence in highly specific non-attenuated oncolytic agents. Here, the scFv to HER2 was inserted in gB (gBHER2). The insertion re-targeted the virus tropism to the HER2-positive cancer cells. This was unexpected since gB is known to be a fusogenic glycoprotein, not a tropism determinant. The gB-retargeted recombinant offered the possibility to investigate how HER2 mediated entry. In contrast to wt-gB, the activation of the chimeric gBHER2 did not require the activation of the gD and of gH/gL by their respective receptors. Furthermore, a soluble form of HER2 could replace the membrane-bound HER2 in mediating virus entry, hinting that HER2 acted by inducing conformational changes to the chimeric gB. This study shows that (i) gB can be modified and become the major determinant of HSV tropism; (ii) the chimeric gBHER2 bypasses the requirement for receptor-mediated activation of other essential entry glycoproteins.


Assuntos
Glicoproteínas/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Receptor ErbB-2/metabolismo , Anticorpos de Cadeia Única/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Glicoproteínas/genética , Herpes Simples/patologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Humanos , Integrinas/genética , Integrinas/metabolismo , Ligantes , Macrolídeos/farmacologia , Nectinas , Receptor ErbB-2/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão , Anticorpos de Cadeia Única/metabolismo , Tropismo Viral , Internalização do Vírus
7.
J Virol ; 91(10)2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28250120

RESUMO

The oncolytic herpes simplex virus (HSV) that has been approved for clinical practice and those HSVs in clinical trials are attenuated viruses, often with the neurovirulence gene γ134.5 and additional genes deleted. One strategy to engineer nonattenuated oncolytic HSVs consists of retargeting the viral tropism to a cancer-specific receptor of choice, exemplified by HER2 (human epidermal growth factor receptor 2), which is present in breast, ovary, and other cancers, and in detargeting from the natural receptors. Because the HER2-retargeted HSVs strictly depend on this receptor for infection, the viruses employed in preclinical studies were cultivated in HER2-positive cancer cells. The production of clinical-grade viruses destined for humans should avoid the use of cancer cells. Here, we engineered the R-213 recombinant, by insertion of a 20-amino-acid (aa) short peptide (named GCN4) in the gH of R-LM113; this recombinant was retargeted to HER2 through insertion in gD of a single-chain antibody (scFv) to HER2. Next, we generated a Vero cell line expressing an artificial receptor (GCN4R) whose N terminus consists of an scFv to GCN4 and therefore is capable of interacting with GCN4 present in gH of R-213. R-213 replicated as well as R-LM113 in SK-OV-3 cells, implying that addition of the GCN4 peptide was not detrimental to gH. R-213 grew to relatively high titers in Vero-GCN4R cells, efficiently spread from cell to cell, and killed both Vero-GCN4R and SK-OV-3 cells, as expected for an oncolytic virus. Altogether, Vero-GCN4R cells represent an efficient system for cultivation of retargeted oncolytic HSVs in non-cancer cells.IMPORTANCE There is growing interest in viruses as oncolytic agents, which can be administered in combination with immunotherapeutic compounds, including immune checkpoint inhibitors. The oncolytic HSV approved for clinical practice and those in clinical trials are attenuated viruses. An alternative to attenuation is a cancer specificity achieved by tropism retargeting to selected cancer receptors. However, the retargeted oncolytic HSVs strictly depend on cancer receptors for infection. Here, we devised a strategy for in vitro cultivation of retargeted HSVs in non-cancer cells. The strategy envisions a double-retargeting approach: one retargeting is via gD to the cancer receptor, and the second retargeting is via gH to an artificial receptor expressed in Vero cells. The double-retargeted HSV uses alternatively the two receptors to infect cancer cells or producer cells. A universal non-cancer cell line for growth of clinical-grade retargeted HSVs represents a step forward in the translational phase.


Assuntos
Vírus Oncolíticos/crescimento & desenvolvimento , Vírus Oncolíticos/genética , Receptor ErbB-2/genética , Simplexvirus/crescimento & desenvolvimento , Cultura de Vírus/métodos , Animais , Linhagem Celular , Chlorocebus aethiops , Engenharia Genética/métodos , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Humanos , Terapia Viral Oncolítica , Vírus Oncolíticos/metabolismo , Receptor ErbB-2/química , Simplexvirus/genética , Simplexvirus/metabolismo , Células Vero , Tropismo Viral
8.
Proc Natl Acad Sci U S A ; 112(29): E3901-10, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26157134

RESUMO

Herpes simplex virus (HSV) is an important human pathogen. It enters cells through an orchestrated process that requires four essential glycoproteins, gD, gH/gL, and gB, activated in cascade fashion by receptor-binding and signaling. gH/gL heterodimer is conserved across the Herpesviridae family. HSV entry is enabled by gH/gL interaction with αvß6- or αvß8-integrin receptors. We report that the interaction of virion gH/gL with integrins resulted in gL dissociation and its release in the medium. gL dissociation occurred if all components of the entry apparatus-receptor-bound gD and gB-were present and was prevented if entry was blocked by a neutralizing monoclonal antibody to gH or by a mutation in gH. We propose that (i) gL dissociation from gH/gL is part of the activation of HSV glycoproteins, critical for HSV entry; and (ii) gL is a functional inhibitor of gH and maintains gH in an inhibited form until receptor-bound gD and integrins signal to gH/gL.


Assuntos
Antígenos de Neoplasias/metabolismo , Herpesvirus Humano 1/fisiologia , Integrinas/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Motivos de Aminoácidos , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Antígenos de Neoplasias/química , Brefeldina A/farmacologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Endocitose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Humanos , Integrinas/química , Microscopia de Fluorescência , Modelos Biológicos , Mutação/genética , Nectinas , Estrutura Terciária de Proteína , Proteólise/efeitos dos fármacos , Solubilidade , Vírion/efeitos dos fármacos , Vírion/metabolismo , Internalização do Vírus/efeitos dos fármacos
9.
J Virol ; 90(8): 4243-4248, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26842473

RESUMO

We report that αvß3 integrin strongly affects the innate immune response in epithelial cells. αvß3 integrin greatly increased the response elicited via plasma membrane Toll-like receptors (TLRs) by herpes simplex virus or bacterial ligands. The endosomal TLR3, not the cytosolic sensor interferon gamma-inducible protein 16 (IFI16), was also boosted by αvß3 integrin. The boosting was exerted specifically by αvß3 integrin but not by αvß6 or αvß8 integrin. Current and previous work indicates that integrin-TLR cooperation occurs in epithelial and monocytic cells. The TLR response should be considered an integrin-TLR response.


Assuntos
Células Epiteliais/metabolismo , Integrina alfaVbeta3/metabolismo , Receptor 3 Toll-Like/metabolismo , Membrana Celular/metabolismo , Endossomos/metabolismo , Inativação Gênica , Células HEK293 , Humanos , Imunidade Inata , Integrina alfaVbeta3/genética , NF-kappa B/metabolismo , Transdução de Sinais , Simplexvirus/metabolismo
10.
PLoS Pathog ; 11(5): e1004907, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25996983

RESUMO

Herpes simplex virus (HSV) enters cells by means of four essential glycoproteins - gD, gH/gL, gB, activated in a cascade fashion by gD binding to one of its receptors, nectin1 and HVEM. We report that the engineering in gH of a heterologous ligand - a single-chain antibody (scFv) to the cancer-specific HER2 receptor - expands the HSV tropism to cells which express HER2 as the sole receptor. The significance of this finding is twofold. It impacts on our understanding of HSV entry mechanism and the design of retargeted oncolytic-HSVs. Specifically, entry of the recombinant viruses carrying the scFv-HER2-gH chimera into HER2+ cells occurred in the absence of gD receptors, or upon deletion of key residues in gD that constitute the nectin1/HVEM binding sites. In essence, the scFv in gH substituted for gD-mediated activation and rendered a functional gD non-essential for entry via HER2. The activation of the gH moiety in the chimera was carried out by the scFv in cis, not in trans as it occurs with wt-gD. With respect to the design of oncolytic-HSVs, previous retargeting strategies were based exclusively on insertion in gD of ligands to cancer-specific receptors. The current findings show that (i) gH accepts a heterologous ligand. The viruses retargeted via gH (ii) do not require the gD-dependent activation, and (iii) replicate and kill cells at high efficiency. Thus, gH represents an additional tool for the design of fully-virulent oncolytic-HSVs retargeted to cancer receptors and detargeted from gD receptors.


Assuntos
DNA Recombinante/metabolismo , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Simplexvirus/fisiologia , Anticorpos de Cadeia Única/metabolismo , Proteínas do Envelope Viral/metabolismo , Tropismo Viral , Internalização do Vírus , Animais , Sítios de Ligação , Linhagem Celular , Sobrevivência Celular , DNA Recombinante/química , DNA Recombinante/uso terapêutico , Terapia Genética , Humanos , Ligantes , Mutação , Neoplasias/metabolismo , Neoplasias/terapia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/uso terapêutico , Engenharia de Proteínas , Receptor ErbB-2/química , Receptor ErbB-2/genética , Receptor ErbB-2/uso terapêutico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Transdução de Sinais , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/uso terapêutico , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/uso terapêutico
11.
PLoS Pathog ; 10(11): e1004477, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25375272

RESUMO

TLR2 is a cell surface receptor which elicits an immediate response to a wide repertoire of bacteria and viruses. Its response is usually thought to be proinflammatory rather than an antiviral. In monocytic cells TLR2 cooperates with coreceptors, e.g. CD14, CD36 and αMß2-integrin. In an earlier work we showed that αvß3-integrin acts in concert with TLR2 to elicit an innate response to HSV, and to lipopolysaccharide. This response is characterized by production of IFN-α and -ß, a specific set of cytokines, and NF-κB activation. We investigated the basis of the cooperation between αvß3-integrin and TLR2. We report that ß3-integrin participates by signaling through Y residues located in the C-tail, known to be involved in signaling activity. αvß3-integrin boosts the MYD88-dependent TLR2 signaling and IRAK4 phosphorylation in 293T and in epithelial, keratinocytic and neuronal cell lines. The replication of ICP0minus HSV is greatly enhanced by DN versions of MYD88, of Akt - a hub of this pathway, or by ß3integrin-silencing. αvß3-integrin enables the recruitment of TLR2, MAL, MYD88 at lipid rafts, the platforms from where the signaling starts. The PAMP of the HSV-induced innate response is the gH/gL virion glycoprotein, which interacts with αvß3-integrin and TLR2 independently one of the other, and cross-links the two receptors. Given the preferential distribution of αvß3-integrin to epithelial cells, we propose that αvß3-integrin serves as coreceptor of TLR2 in these cells. The results open the possibility that TLR2 makes use of coreceptors in a variety of cells to broaden its spectrum of activity and tissue specificity.


Assuntos
Bactérias/imunologia , Integrina alfaVbeta3/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia , Simplexvirus/imunologia , Receptor 2 Toll-Like/imunologia , Bactérias/genética , Células HeLa , Herpes Simples/genética , Herpes Simples/imunologia , Humanos , Integrina alfaVbeta3/genética , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/imunologia , Queratinócitos/imunologia , Fator 88 de Diferenciação Mieloide/genética , Neurônios/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Transdução de Sinais/genética , Simplexvirus/genética , Receptor 2 Toll-Like/genética
12.
PLoS Pathog ; 9(12): e1003806, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24367260

RESUMO

Herpes simplex virus (HSV)--and herpesviruses in general--encode for a multipartite entry/fusion apparatus. In HSV it consists of the HSV-specific glycoprotein D (gD), and three additional glycoproteins, gH/gL and gB, conserved across the Herpesviridae family and responsible for the execution of fusion. According to the current model, upon receptor binding, gD propagates the activation to gH/gL and to gB in a cascade fashion. Questions remain about how the cascade of activation is controlled and how it is synchronized with virion endocytosis, to avoid premature activation and exhaustion of the glycoproteins. We considered the possibility that such control might be carried out by as yet unknown receptors. Indeed, receptors for HSV gB, but not for gH/gL, have been described. In other members of the Herpesviridae family, such as Epstein-Barr virus, integrin receptors bind gH/gL and trigger conformational changes in the glycoproteins. We report that αvß6- and αvß8-integrins serve as receptors for HSV entry into experimental models of keratinocytes and other epithelial and neuronal cells. Evidence rests on loss of function experiments, in which integrins were blocked by antibodies or silenced, and gain of function experiments in which αvß6-integrin was expressed in integrin-negative cells. αvß6- and αvß8-integrins acted independently and are thus interchangeable. Both bind gH/gL with high affinity. The interaction profoundly affects the route of HSV entry and directs the virus to acidic endosomes. In the case of αvß8, but not αvß6-integrin, the portal of entry is located at lipid microdomains and requires dynamin 2. Thus, a major role of αvß6- or αvß8-integrin in HSV infection appears to be to function as gH/gL receptors and to promote virus endocytosis. We propose that placing the gH/gL activation under the integrin trigger point enables HSV to synchronize virion endocytosis with the cascade of glycoprotein activation that culminates in execution of fusion.


Assuntos
Antígenos de Neoplasias/fisiologia , Integrinas/fisiologia , Receptores Virais , Simplexvirus/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Células Cultivadas , Endocitose/fisiologia , Células HEK293 , Células HeLa , Humanos , Células K562 , Receptores Virais/metabolismo , Células Sf9 , Spodoptera
13.
PLoS Pathog ; 9(1): e1003155, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23382683

RESUMO

Oncolytic viruses aim to specifically kill tumor cells. A major challenge is the effective targeting of disseminated tumors in vivo. We retargeted herpes simplex virus (HSV) tropism to HER-2 oncoprotein p185, overexpressed in ovary and breast cancers. The HER-2-retargeted R-LM249 exclusively infects and kills tumor cells expressing high levels of human HER-2. Here, we assessed the efficacy of systemically i.p. delivered R-LM249 against disseminated tumors in mouse models that recapitulate tumor spread to the peritoneum in women. The human ovarian carcinoma SK-OV-3 cells implanted intraperitoneally (i.p.) in immunodeficient Rag2⁻/⁻;Il2rg⁻/⁻ mice gave rise to a progressive peritoneal carcinomatosis which mimics the fatal condition in advanced human patients. I.p. administration of R-LM249 strongly inhibited carcinomatosis, resulting in 60% of mice free from peritoneal diffusion, and 95% reduction in the total weight of neoplastic nodules. Intraperitoneal metastases are a common outcome in breast cancer: i.p. administration of R-LM249 strongly inhibited the growth of ovarian metastases of HER-2+ MDA-MB-453 breast cells. Brain metastases were also reduced. Cumulatively, upon i.p. administration the HER-2-redirected oncolytic HSV effectively reduced the growth of ovarian and breast carcinoma disseminated to the peritoneal cavity.


Assuntos
Neoplasias da Mama/terapia , Herpesvirus Humano 1/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Neoplasias Ovarianas/terapia , Neoplasias Peritoneais/terapia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Linhagem Celular Tumoral , Sobrevivência Celular , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Herpesvirus Humano 1/fisiologia , Humanos , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Vírus Oncolíticos/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/mortalidade , Neoplasias Peritoneais/mortalidade , Neoplasias Peritoneais/secundário , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Proc Natl Acad Sci U S A ; 109(48): 19792-7, 2012 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-23150579

RESUMO

Pathogens are sensed by Toll-like receptors (TLRs) and a growing number of non-TLR receptors. Integrins constitute a family of signaling receptors exploited by viruses and bacteria to access cells. By gain- and loss-of-function approaches we found that αvß3-integrin is a sensor of and plays a crucial role in the innate defense against herpes simplex virus (HSV). αvß3-integrin signaled through two pathways. One concurred with TLR2, affected activation/induction of interferons type 1 (IFNs-1), NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), and a polarized set of cytokines and receptors. The virion glycoproteins gH/gL sufficed to induce IFN1 and NF-κB via this pathway. The other pathway was TLR2-independent, involved sarcoma (SRC)-spleen tyrosine kinase (SYK)-Caspase recruitment domain-containing protein 9 (CARD9)-TRIF (TIR-domain-containing adapter-inducing interferon-ß), and affected interferon regulatory factor 3 and 7 (IRF3-IRF7). The importance of αvß3-integrin-mediated defense is reflected in the observation that HSV evolved the immediate-early infected cellular protein 0 (ICP0) protein to counteract it. We propose that αvß3-integrin is considered a class of non-TLR pattern recognition receptors, a role likely exerted toward viruses and bacteria that interact with integrins and mount an innate response.


Assuntos
Herpesvirus Humano 1/imunologia , Imunidade Inata/fisiologia , Integrina alfaVbeta3/fisiologia , Linhagem Celular , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Inativação Gênica , Herpesvirus Humano 1/fisiologia , Humanos , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase , Replicação Viral
15.
J Virol ; 87(24): 13911-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109241

RESUMO

αvß3 integrin represents a novel sensing system which detects herpes simplex virus (HSV) and bacterial constituents. In cooperation with Toll-like receptor 2 (TLR2), it elicits an innate response that leads to activation of type I interferon (IFN), NF-κB, and a specific set of cytokines. We report that this defensive branch is functional in cells which represent experimental models of epithelial, including keratinocytic, and neuronal cells. These are the major targets of HSV in vivo. HSV entered the three cell lines via distinct routes. Hence, the defensive response was independent of the route of virus entry. Soluble gH/gL sufficed to elicit type I IFN and NF-κB activation and represents the viral pathogen-associated molecular pattern (PAMP) of this defense system.


Assuntos
Células Epiteliais/imunologia , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Integrina alfa5/imunologia , Integrina beta3/imunologia , Interferon Tipo I/imunologia , Glicoproteínas de Membrana/imunologia , Chaperonas Moleculares/imunologia , NF-kappa B/imunologia , Neurônios/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas Virais/imunologia , Linhagem Celular , Contraindicações , Células Epiteliais/virologia , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Humanos , Integrina beta3/genética , Interferon Tipo I/genética , Glicoproteínas de Membrana/genética , Chaperonas Moleculares/genética , NF-kappa B/genética , Neurônios/virologia , Proteínas do Envelope Viral/genética , Proteínas Virais/genética
16.
17.
Cancers (Basel) ; 16(6)2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38539478

RESUMO

The role of the interaction with cell-surface glycosaminoglycans (GAGs) during in vivo HSV infection is currently unknown. The rationale of the current investigation was to improve the anticancer efficacy of systemically administered retargeted oHSVs (ReHVs) by decreasing their binding to GAGs, including those of endothelial cells, blood cells, and off-tumor tissues. As a proof-of-principle approach, we deleted seven amino acids critical for interacting with GAGs from the glycoprotein C (gC) of R-337 ReHV. The modification in the resulting R-399 recombinant prolonged the half-life in the blood of systemically administered R-399 and enhanced its biodistribution to tumor-positive lungs and to the tumor-negative liver. Ultimately, it greatly increased the R-399 efficacy against metastatic-like lung tumors upon IV administration but not against subcutaneous tumors upon IT administration. These results provide evidence that the increased efficacy seen upon R-399 systemic administration correlated with the slower clearance from the circulation. To our knowledge, this is the first in vivo evidence that the partial impairment of the gC interaction with GAGs resulted in a prolonged half-life of circulating ReHV, an increase in the amount of ReHV taken up by tissues and tumors, and, ultimately, an enhanced anticancer efficacy of systemically administered ReHV.

18.
J Virol ; 86(5): 2850-5, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22171266

RESUMO

Herpes simplex virus (HSV) enters cells by fusion at plasma membranes or endosomes. Cellular factors route the virus to different pathways. αVß3-integrin directs HSV to a lipid raft and acidic endosome pathway. We report that infection mediated by nectin1 plus αVß3-integrin exhibits the same characteristics as entry mediated by raft-located forms of nectin. αVß3-integrin relocalizes nectin1 to lipid rafts, independently of virus. Thus, HSV routing to the lipid raft-dependent pathway is consequent to the integrin-induced relocalization of nectin1. Inhibition by the Na+/H+ exchanger 5-(N-ethyl-N-isopropyl)amirolide suggests that αVß3-integrin overexpression favors HSV macropinocytic uptake in some cells but not in others.


Assuntos
Moléculas de Adesão Celular/metabolismo , Herpes Simples/metabolismo , Herpesvirus Humano 1/fisiologia , Integrina alfaVbeta3/metabolismo , Microdomínios da Membrana/metabolismo , Moléculas de Adesão Celular/genética , Linhagem Celular , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Humanos , Integrina alfaVbeta3/genética , Microdomínios da Membrana/virologia , Nectinas , Transporte Proteico , Internalização do Vírus
19.
J Virol ; 86(12): 6555-62, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22496225

RESUMO

A number of sentinels sense incoming herpes simplex virus (HSV) virions and initiate an immediate innate response. The first line of defense at the cell surface is TLR2 (Toll-like receptor 2), whose signature signaling activity leads to activation of the key transcription factor NF-κB. We report that the HSV pathogen-associated molecular patterns for TLR2 are the virion glycoproteins gH/gL and gB, which constitute the conserved fusion core apparatus across the members of the Herpesviridae family. Specifically, virions devoid singly of one of essential fusion glycoproteins (gD, gB, or gH null), able to attach to cells but defective in fusion/entry, were sufficient to elicit the first wave of NF-κB response to HSV. The most effective were the gD-null virions, positive for gH/gL and gB. A soluble form of gB, truncated upstream of the transmembrane sequence (gB(730t-st)), was produced in human cells and purified by means of a Strep tag. gH/gL and gB were each able to physically interact with TLR2 in coimmunoprecipitation assays, one independently of the other, yet gH(t-st)/gL, but not gB(730t-st), elicited an NF-κB response. Thus, whereas both HSV gH/gL and gB are ligands to TLR2, only gH/gL is sufficient to initiate a signaling cascade which leads to NF-κB activation.


Assuntos
Herpes Simples/metabolismo , Herpesvirus Humano 1/metabolismo , NF-kappa B/metabolismo , Receptor 2 Toll-Like/metabolismo , Proteínas do Envelope Viral/metabolismo , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Humanos , NF-kappa B/genética , Ligação Proteica , Transdução de Sinais , Receptor 2 Toll-Like/genética , Proteínas do Envelope Viral/genética
20.
Mol Ther ; 20(5): 994-1001, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22354378

RESUMO

Oncolytic herpes simplex viruses (HSVs) represent a novel frontier against tumors resistant to standard therapies, like glioblastoma (GBM). The oncolytic HSVs that entered clinical trials so far showed encouraging results; however, they are marred by the fact that they are highly attenuated. We engineered HSVs that maintain unimpaired lytic efficacy and specifically target cells that express tumor-specific receptors, thus limiting the cytotoxicity only to cancer cells, and leaving unharmed the neighboring tissues. We report on the safety and efficacy in a high-grade glioma (HGG) model of R-LM113, an HSV recombinant retargeted to human epidermal growth factor receptor 2 (HER2), frequently expressed in GBMs. We demonstrated that R-LM113 is safe in vivo as it does not cause encephalitis when intracranially injected in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice, extremely sensitive to wild-type HSV. The efficacy of R-LM113 was assessed in a platelet-derived growth factor (PDGF)-induced infiltrative glioma model engineered to express HER2 and transplanted intracranially in adult NOD/SCID mice. Mice injected with HER2-engineered glioma cells infected with R-LM113 showed a doubled survival time compared with mice injected with uninfected cells. A doubling in survival time from the beginning of treatment was obtained also when R-LM113 was administered into already established tumors. These data demonstrate the efficacy of R-LM113 in thwarting tumor growth.


Assuntos
Neoplasias Encefálicas/terapia , Vetores Genéticos , Glioma/terapia , Terapia Viral Oncolítica/métodos , Receptor ErbB-2/genética , Simplexvirus/genética , Adulto , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Encefalite/prevenção & controle , Engenharia Genética , Glioma/genética , Glioma/mortalidade , Humanos , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Gradação de Tumores , Transplante de Neoplasias , Fator de Crescimento Derivado de Plaquetas/genética , Taxa de Sobrevida , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA