Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(40): 20141-20150, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31527243

RESUMEN

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.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263255

RESUMEN

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.


Asunto(s)
Virus Oncolíticos , Péptidos , Proteínas del Envoltorio Viral , Tropismo Viral/genética , Animales , Chlorocebus aethiops , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Péptidos/genética , Péptidos/metabolismo , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
3.
PLoS Pathog ; 13(4): e1006352, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28423057

RESUMEN

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.


Asunto(s)
Glicoproteínas/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Receptor ErbB-2/metabolismo , Anticuerpos de Cadena Única/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Glicoproteínas/genética , Herpes Simple/patología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidad , Humanos , Integrinas/genética , Integrinas/metabolismo , Ligandos , Macrólidos/farmacología , Nectinas , Receptor ErbB-2/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Proteínas Recombinantes de Fusión , Anticuerpos de Cadena Única/metabolismo , Tropismo Viral , Internalización del Virus
4.
Proc Natl Acad Sci U S A ; 112(29): E3901-10, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26157134

RESUMEN

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.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Herpesvirus Humano 1/fisiología , Integrinas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Antígenos de Neoplasias/química , Brefeldino A/farmacología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Endocitosis/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/virología , Herpes Simple/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/efectos de los fármacos , Humanos , Integrinas/química , Microscopía Fluorescente , Modelos Biológicos , Mutación/genética , Nectinas , Estructura Terciaria de Proteína , Proteolisis/efectos de los fármacos , Solubilidad , Virión/efectos de los fármacos , Virión/metabolismo , Internalización del Virus/efectos de los fármacos
5.
J Virol ; 90(8): 4243-4248, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26842473

RESUMEN

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.


Asunto(s)
Células Epiteliales/metabolismo , Integrina alfaVbeta3/metabolismo , Receptor Toll-Like 3/metabolismo , Membrana Celular/metabolismo , Endosomas/metabolismo , Silenciador del Gen , Células HEK293 , Humanos , Inmunidad Innata , Integrina alfaVbeta3/genética , FN-kappa B/metabolismo , Transducción de Señal , Simplexvirus/metabolismo
6.
PLoS Pathog ; 10(11): e1004477, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375272

RESUMEN

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.


Asunto(s)
Bacterias/inmunología , Integrina alfaVbeta3/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/inmunología , Simplexvirus/inmunología , Receptor Toll-Like 2/inmunología , Bacterias/genética , Células HeLa , Herpes Simple/genética , Herpes Simple/inmunología , Humanos , Integrina alfaVbeta3/genética , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Queratinocitos/inmunología , Factor 88 de Diferenciación Mieloide/genética , Neuronas/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Transducción de Señal/genética , Simplexvirus/genética , Receptor Toll-Like 2/genética
7.
PLoS Pathog ; 9(12): e1003806, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367260

RESUMEN

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.


Asunto(s)
Antígenos de Neoplasias/fisiología , Integrinas/fisiología , Receptores Virales , Simplexvirus/fisiología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Células Cultivadas , Endocitosis/fisiología , Células HEK293 , Células HeLa , Humanos , Células K562 , Receptores Virales/metabolismo , Células Sf9 , Spodoptera
8.
Proc Natl Acad Sci U S A ; 109(48): 19792-7, 2012 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-23150579

RESUMEN

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.


Asunto(s)
Herpesvirus Humano 1/inmunología , Inmunidad Innata/fisiología , Integrina alfaVbeta3/fisiología , Línea Celular , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Silenciador del Gen , Herpesvirus Humano 1/fisiología , Humanos , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa , Replicación Viral
9.
J Virol ; 87(24): 13911-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109241

RESUMEN

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.


Asunto(s)
Células Epiteliales/inmunología , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Integrina alfa5/inmunología , Integrina beta3/inmunología , Interferón Tipo I/inmunología , Glicoproteínas de Membrana/inmunología , Chaperonas Moleculares/inmunología , FN-kappa B/inmunología , Neuronas/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/inmunología , Línea Celular , Contraindicaciones , Células Epiteliales/virología , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Humanos , Integrina beta3/genética , Interferón Tipo I/genética , Glicoproteínas de Membrana/genética , Chaperonas Moleculares/genética , FN-kappa B/genética , Neuronas/virología , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética
10.
Cancers (Basel) ; 16(6)2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38539478

RESUMEN

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.

11.
Front Mol Biosci ; 11: 1444446, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38983248

RESUMEN

[This corrects the article DOI: 10.3389/fmolb.2023.1149973.].

12.
J Virol ; 86(5): 2850-5, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22171266

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Herpes Simple/metabolismo , Herpesvirus Humano 1/fisiología , Integrina alfaVbeta3/metabolismo , Microdominios de Membrana/metabolismo , Moléculas de Adhesión Celular/genética , Línea Celular , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Humanos , Integrina alfaVbeta3/genética , Microdominios de Membrana/virología , Nectinas , Transporte de Proteínas , Internalización del Virus
13.
J Virol ; 86(12): 6555-62, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22496225

RESUMEN

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.


Asunto(s)
Herpes Simple/metabolismo , Herpesvirus Humano 1/metabolismo , FN-kappa B/metabolismo , Receptor Toll-Like 2/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Humanos , FN-kappa B/genética , Unión Proteica , Transducción de Señal , Receptor Toll-Like 2/genética , Proteínas del Envoltorio Viral/genética
14.
Proc Natl Acad Sci U S A ; 107(51): 22260-5, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21135248

RESUMEN

HSVs enter cells in a receptor-dependent [nectin1 or herpesviruses entry mediator (HVEM)] fashion by fusion of the viral envelope with plasma membrane (neutral pH compartment), by endocytosis into neutral or acidic compartments, or by macropinocytosis/phagocytosis. The cellular determinants of the route of entry are unknown. Here, we asked what cellular factors determine the pathway of HSV entry. CHO cells lack ß(3)-integrin and the respective α-subunits' heterodimers. We report that, in the absence of α(V)ß(3)-integrin, HSV enters CHO-nectin1 cells through a pathway independent of cholesterol-rich rafts and dynamin2. In the presence of α(V)ß(3)-integrin, HSV enters CHO-nectin1 cells through a pathway dependent on cholesterol-rich rafts and dynamin2. HSV enters J-nectin1 and 293T cells through a neutral compartment independent of cholesterol-rich rafts and dynamin2. α(V)ß(3)-integrin overexpression in these cells modifies the route of entry to an acidic compartment dependent on cholesterol-rich rafts and dynamin2, hence similar to that in α(V)ß(3)-integrin-positive CHO-nectin1 cells. In some cells, the diversion of entry from an integrin- and raft-independent pathway to an acidic compartment requiring cholesterol-rich lipids rafts and dynamin2 is irreversible. Indeed, HSV cannot infect CHO-nectin1-α(V)ß(3) cells through any compartment when the αvß3-integrin-dependent pathway is blocked by anti-integrin antibody, anti-dynamin2, or anti-acidification drugs. We conclude that the αvß3-integrin is a determinant in the choice of HSV entry pathway into cells. Because the pathway dictated by αvß3-integrin is through lipid rafts, the platforms for a number of Toll-like receptors, current findings raise the possibility that αvß3-integrin acts as a sentinel of innate immunity.


Asunto(s)
Dinamina II/metabolismo , Integrina alfaVbeta3/metabolismo , Microdominios de Membrana/metabolismo , Simplexvirus/fisiología , Internalización del Virus , Animales , Anticuerpos/farmacología , Células CHO , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Colesterol/genética , Colesterol/inmunología , Colesterol/metabolismo , Cricetinae , Cricetulus , Dinamina II/genética , Dinamina II/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/fisiología , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/inmunología , Microdominios de Membrana/genética , Microdominios de Membrana/inmunología , Nectinas
15.
Front Mol Biosci ; 10: 1149973, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37251078

RESUMEN

Nectin4 is a recently discovered tumor associated antigen expressed in cancers that constitute relevant unmet clinical needs, including the undruggable triple negative breast cancer, pancreatic ductal carcinoma, bladder/urothelial cancer, cervical cancer, lung carcinoma and melanoma. So far, only one nectin4-specific drug-Enfortumab Vedotin-has been approved and the clinical trials that test novel therapeutics are only five. Here we engineered R-421, an innovative retargeted onco-immunotherapeutic herpesvirus highly specific for nectin4 and unable to infect through the natural herpes receptors, nectin1 or herpesvirus entry mediator. In vitro, R-421 infected and killed human nectin4-positive malignant cells and spared normal cells, e.g., human fibroblasts. Importantly from a safety viewpoint, R-421 failed to infect malignant cells that do not harbor nectin4 gene amplification/overexpression, whose expression level was moderate-to-low. In essence, there was a net threshold value below which cells were spared from infection, irrespective of whether they were malignant or normal; the only cells that R-421 targeted were the malignant overexpressing ones. In vivo, R-421 decreased or abolished the growth of murine tumors made transgenic for human nectin4 and conferred sensitivity to immune checkpoint inhibitors in combination therapies. Its efficacy was augmented by the cyclophosphamide immunomodulator and decreased by depletion of CD8-positive lymphocytes, arguing that it was in part T cell-mediated. R-421 elicited in-situ vaccination that protected from distant challenge tumors. This study provides proof-of-principle specificity and efficacy data justifying nectin4-retargeted onco-immunotherapeutic herpesvirus as an innovative approach against a number of difficult-to-drug clinical indications.

16.
Cancers (Basel) ; 15(16)2023 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-37627072

RESUMEN

We investigated the anticancer efficacy, blood clearance, and tissue biodistribution of systemically administered retargeted oncolytic herpes simplex viruses (ReHVs) in HSV-naïve and HSV-preimmunized (HSV-IMM) mice. Efficacy was tested against lung tumors formed upon intravenous administration of cancer cells, a model of metastatic disease, and against subcutaneous distant tumors. In naïve mice, HER2- and hPSMA-retargeted viruses, both armed with mIL-12, were highly effective, even when administered to mice with well-developed tumors. Efficacy was higher for combination regimens with immune checkpoint inhibitors. A significant amount of infectious virus persisted in the blood for at least 1 h. Viral genomes, or fragments thereof, persisted in the blood and tissues for days. Remarkably, the only sites of viral replication were the lungs of tumor-positive mice and the subcutaneous tumors. No replication was detected in other tissues, strengthening the evidence of the high cancer specificity of ReHVs, a property that renders ReHVs suitable for systemic administration. In HSV-IMM mice, ReHVs administered at late times failed to exert anticancer efficacy, and the circulating virus was rapidly inactivated. Serum stability and in vivo whole blood stability assays highlighted neutralizing antibodies as the main factor in virus inactivation. Efforts to deplete mice of the neutralizing antibodies are ongoing.

17.
J Virol ; 84(8): 4013-25, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20147400

RESUMEN

Herpes simplex virus (HSV) fusion with cells requires the gD, gB, and gH/gL glycoprotein quartet. gD serves as a receptor binding glycoprotein. gB and gH/gL execute fusion in an as-yet-unclear manner. To better understand the role of gH/gL in HSV entry, we produced a soluble version of gH/gL carrying a One-STrEP tag (gH(t.st)/gL). Previous findings implicated integrins as possible ligands to gH/gL (C. Parry et al., J. Gen. Virol. 86:7-10, 2005). We report that (i) gH(t.st)/gL bound a number of cells in a dose-dependent manner at concentrations similar to those required for the binding of soluble gB or gD. (ii) gH(t.st)/gL inhibited HSV entry at the same concentrations required for binding. It also inhibited cell-cell fusion in transfected cells. (iii) The absence of beta3 integrin did not prevent the binding of gH(t.st)/gL to CHO cells and infection inhibition. Conversely, integrin-negative K562 cells did not acquire the ability to bind gH(t.st)/gL when hyperexpressing alphaVbeta3 integrin. (iv) Constitutive expression of wild-type gH/gL (wt-gH/gL) restricted infection in all of the cell lines tested, a behavior typical of glycoproteins which bind cellular receptors. The extent of restriction broadly paralleled the efficiency of gH/gL transfection. RGD motif mutant gH/gL could not be differentiated from wt-gH with respect to restriction of infection. Cumulatively, the present results provide several lines of evidence that HSV gH/gL interacts with a cell surface cognate protein(s), that this protein is not necessarily an alphaVbeta3 integrin, and that this interaction is required for the process of virus entry/fusion.


Asunto(s)
Simplexvirus/fisiología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Línea Celular , Humanos , Integrina beta3/metabolismo , Unión Proteica
18.
Viruses ; 13(10)2021 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-34696515

RESUMEN

Prostate specific membrane antigen (PSMA) is a specific high frequency cell surface marker of prostate cancers. Theranostic approaches targeting PSMA show no major adverse effects and rule out off-tumor toxicity. A PSMA-retargeted oHSV (R-405) was generated which both infected and was cytotoxic exclusively for PSMA-positive cells, including human prostate cancer LNCaP and 22Rv1 cells, and spared PSMA-negative cells. R-405 in vivo efficacy against LLC1-PSMA and Renca-PSMA tumors consisted of inhibiting primary tumor growth, establishing long-term T immune response, immune heating of the microenvironment, de-repression of the anti-tumor immune phenotype, and sensitization to checkpoint blockade. The in situ vaccination protected from distant challenge tumors, both PSMA-positive and PSMA-negative, implying that it was addressed also to LLC1 tumor antigens. PSMA-retargeted oHSVs are a precision medicine tool worth being additionally investigated in the immunotherapeutic and in situ vaccination landscape against prostate cancers.


Asunto(s)
Viroterapia Oncolítica/métodos , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/terapia , Antineoplásicos/metabolismo , Biomarcadores/sangre , Línea Celular Tumoral , Humanos , Hibridación in Situ/métodos , Calicreínas/inmunología , Masculino , Virus Oncolíticos/genética , Medicina de Precisión/métodos , Microambiente Tumoral , Vacunación/métodos
19.
Cancers (Basel) ; 13(2)2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33445744

RESUMEN

Our laboratory has pursued the generation of cancer-specific oncolytic herpes simplex viruses (oHSVs) which ensure high efficacy while maintaining a high safety profile. Their blueprint included retargeting to a Tumor-Associated Antigen, e.g., HER2, coupled to detargeting from natural receptors to avoid off-target and off-tumor infections and preservation of the full complement of unmodified viral genes. These oHSVs are "fully virulent in their target cancer cells". The 3rd generation retargeted oHSVs carry two distinct retargeting moieties, which enable infection of a producer cell line and of the target cancer cells, respectively. They can be propagated in an ad hoc Vero cell derivative at about tenfold higher yields than 1st generation recombinants, and more effectively replicate in human cancer cell lines. The R-335 and R-337 prototypes were armed with murine IL-12. Intratumorally-administered R-337 conferred almost complete protection from LLC-1-HER2 primary tumors, unleashed the tumor microenvironment immunosuppression, synergized with the checkpoint blockade and conferred long-term vaccination against distant challenge tumors. In summary, the problem intrinsic to the propagation of retargeted oHSVs-which strictly require cells positive for targeted receptors-was solved in 3rd generation viruses. They are effective as immunotherapeutic agents against primary tumors and as antigen-agnostic vaccines.

20.
Viruses ; 13(9)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34578328

RESUMEN

We report on the efficacy of the non-attenuated HER2-retargeted oHSV named R-337 against the immunologically hot CT26-HER2 tumor, and an insight into the basis of the immune protection. Preliminarily, we conducted an RNA immune profiling and immune cell content characterization of CT26-HER2 tumor in comparison to the immunologically cold LLC1-HER2 tumor. CT26-HER2 tumor was implanted into HER2-transgenic BALB/c mice. Hallmarks of R-337 effects were the protection from primary tumor, long-term adaptive vaccination directed to both HER2 and CT26-wt cell neoantigens. The latter effect differentiated R-337 from OncoVEXGM-CSF. As to the basis of the immune protection, R-337 orchestrated several changes to the tumor immune profile, which cumulatively reversed the immunosuppression typical of this tumor (graphical abstract). Thus, Ido1 (inhibitor of T cell anticancer immunity) levels and T regulatory cell infiltration were decreased; Cd40 and Cd27 co-immunostimulatory markers were increased; the IFNγ cascade was activated. Of note was the dampening of IFN-I response, which we attribute to the fact that R-337 is fully equipped with genes that contrast the host innate response. The IFN-I shut-down likely favored viral replication and the expression of the mIL-12 payload, which, in turn, boosted the antitumor response. The results call for a characterization of tumor immune markers to employ oncolytic herpesviruses more precisely.


Asunto(s)
Genotipo , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Inmunidad , Inmunoterapia/métodos , Neoplasias/inmunología , Virus Oncolíticos/inmunología , Animales , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Virus Oncolíticos/patogenicidad , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA