Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(1): e1008961, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411789

RESUMO

Varicella-zoster virus (VZV) is a medically important alphaherpesvirus that induces fusion of the virion envelope and the cell membrane during entry, and between cells to form polykaryocytes within infected tissues during pathogenesis. All members of the Herpesviridae, including VZV, have a conserved core fusion complex composed of glycoproteins, gB, gH and gL. The ectodomain of the primary fusogen, gB, has five domains, DI-V, of which DI contains the fusion loops needed for fusion function. We recently demonstrated that DIV is critical for fusion initiation, which was revealed by a 2.8Å structure of a VZV neutralizing mAb, 93k, bound to gB and mutagenesis of the gB-93k interface. To further assess the mechanism of mAb 93k neutralization, the binding site of a non-neutralizing mAb to gB, SG2, was compared to mAb 93k using single particle cryogenic electron microscopy (cryo-EM). The gB-SG2 interface partially overlapped with that of gB-93k but, unlike mAb 93k, mAb SG2 did not interact with the gB N-terminus, suggesting a potential role for the gB N-terminus in membrane fusion. The gB ectodomain structure in the absence of antibody was defined at near atomic resolution by single particle cryo-EM (3.9Å) of native, full-length gB purified from infected cells and by X-ray crystallography (2.4Å) of the transiently expressed ectodomain. Both structures revealed that the VZV gB N-terminus (aa72-114) was flexible based on the absence of visible structures in the cryo-EM or X-ray crystallography data but the presence of gB N-terminal peptides were confirmed by mass spectrometry. Notably, N-terminal residues 109KSQD112 were predicted to form a small α-helix and alanine substitution of these residues abolished cell-cell fusion in a virus-free assay. Importantly, transferring the 109AAAA112 mutation into the VZV genome significantly impaired viral propagation. These data establish a functional role for the gB N-terminus in membrane fusion broadly relevant to the Herpesviridae.


Assuntos
Herpesvirus Humano 3/fisiologia , Melanoma/metabolismo , Fusão de Membrana , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Melanoma/virologia , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Homologia de Sequência , Células Tumorais Cultivadas , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
2.
J Immunol ; 204(11): 2961-2972, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32284333

RESUMO

CMV has been proposed to play a role in cancer progression and invasiveness. However, CMV has been increasingly studied as a cancer vaccine vector, and multiple groups, including ours, have reported that the virus can drive antitumor immunity in certain models. Our previous work revealed that intratumoral injections of wild-type murine CMV (MCMV) into B16-F0 melanomas caused tumor growth delay in part by using a viral chemokine to recruit macrophages that were subsequently infected. We now show that MCMV acts as a STING agonist in the tumor. MCMV infection of tumors in STING-deficient mice resulted in normal recruitment of macrophages to the tumor, but poor recruitment of CD8+ T cells, reduced production of inflammatory cytokines and chemokines, and no delay in tumor growth. In vitro, expression of type I IFN was dependent on both STING and the type I IFNR. Moreover, type I IFN alone was sufficient to induce cytokine and chemokine production by macrophages and B16 tumor cells, suggesting that the major role for STING activation was to produce type I IFN. Critically, viral infection of wild-type macrophages alone was sufficient to restore tumor growth delay in STING-deficient animals. Overall, these data show that MCMV infection and sensing in tumor-associated macrophages through STING signaling is sufficient to promote antitumor immune responses in the B16-F0 melanoma model.


Assuntos
Infecções por Herpesviridae/imunologia , Melanoma/imunologia , Proteínas de Membrana/metabolismo , Muromegalovirus/fisiologia , Neoplasias Cutâneas/imunologia , Macrófagos Associados a Tumor/imunologia , Animais , Movimento Celular , Modelos Animais de Doenças , Humanos , Imunidade/genética , Interferon Tipo I/metabolismo , Melanoma/virologia , Melanoma Experimental , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Cutâneas/virologia , Carga Tumoral , Microambiente Tumoral
3.
Br J Cancer ; 124(8): 1357-1360, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33558707

RESUMO

The COVID-19 pandemic has had an unprecedented impact on the National Health Service in United Kingdom. The UK Ocular Oncology Services evaluated the impact on the adult eye cancer care in the UK. All four adult Ocular Oncology centres participated in a multicentre retrospective review comparing uveal melanoma referral patterns and treatments in a 4-month period during the national lockdown and first wave of the COVID-19 pandemic in 2020 with corresponding periods in previous 2 years. During the national lockdown, referral numbers and confirmed uveal melanoma cases reduced considerably, equalling to ~120 fewer diagnosed uveal melanoma cases compared to previous 2 years. Contrary to the recent trend, increased caseloads of enucleation and stereotactic radiosurgery (p > 0.05), in comparison to fewer proton beam therapy (p < 0.05), were performed. In the 4-month period following lockdown, there was a surge in clinical activities with more advanced diseases (p < 0.05) presenting to the services. As the COVID-19 pandemic continues to mount pressure and reveal its hidden impact on the eye cancer care, it is imperative for the Ocular Oncology Services to plan recovery strategies and innovative ways of working.


Assuntos
COVID-19/epidemiologia , Neoplasias Oculares/epidemiologia , Melanoma/epidemiologia , Pandemias , Neoplasias Uveais/epidemiologia , COVID-19/complicações , COVID-19/terapia , COVID-19/virologia , Controle de Doenças Transmissíveis/métodos , Neoplasias Oculares/complicações , Neoplasias Oculares/terapia , Neoplasias Oculares/virologia , Humanos , Melanoma/complicações , Melanoma/terapia , Melanoma/virologia , Terapia com Prótons/métodos , SARS-CoV-2/patogenicidade , Medicina Estatal , Reino Unido/epidemiologia , Neoplasias Uveais/complicações , Neoplasias Uveais/terapia , Neoplasias Uveais/virologia
4.
Exp Eye Res ; 202: 108285, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33039456

RESUMO

Uveal melanoma (UM), the most common primary malignant tumor of the eye in adults, is difficult-to-treat. UM has a relatively high mortality secondary to distant metastasis and poor overall survival with existing therapies. The oncolytic virus herpes simplex virus type-1 (HSV-1) has been approved for clinical use in melanoma. This double-stranded DNA virus was suspected to directly activate lysis specifically in neoplastic cells. We tested the antitumor efficacy of recombinant oncolytic HSV-1-EGFP (oHSV-EGFP) in UM and characterized the local and systemic antitumor innate immune response in a murine xenograft model. We first determined the efficacy of the oncolytic virus in 92.1, MUM2B and MP41 cell lines. In murine xenograft models, oHSV-EGFP reduced intraocular tumors as well as systemic subcutaneous tumors. A significant change in cytokines was observed in viral infected cells, especially a rise in IFNγ. In vivo analyses showed increased anti-tumorigenic M1 macrophages and decreased pro-tumorigenic M2 macrophages in peripheral blood, and intraocular and distant tumors after intravitreal viral treatment. Increased infiltration of natural killer cells and mature dendritic cells was also detected after viral treatment. In addition, no virus was detected in major organs after the treatment. Our data support that oHSV-EGFP is effective, neoplasm specific, immune active and safe, providing possible clinical translatable options to treat ocular and metastatic UM.


Assuntos
Modelos Animais de Doenças , Proteínas de Fluorescência Verde/fisiologia , Herpesvirus Humano 1/fisiologia , Ativação de Macrófagos/fisiologia , Melanoma/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Neoplasias Uveais/terapia , Animais , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Xenoenxertos , Humanos , Melanoma/metabolismo , Melanoma/patologia , Melanoma/virologia , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias Uveais/metabolismo , Neoplasias Uveais/patologia , Neoplasias Uveais/virologia
5.
J Med Virol ; 92(10): 2081-2086, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32374427

RESUMO

SARS-CoV-2 is assumed to use angiotensin-converting enzyme 2 (ACE2) and other auxiliary proteins for cell entry. Recent studies have described conjunctival congestion in 0.8% of patients with laboratory-confirmed severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), and there has been speculation that SARS-CoV-2 can be transmitted through the conjunctiva. However, it is currently unclear whether conjunctival epithelial cells express ACE2 and its cofactors. In this study, a total of 38 conjunctival samples from 38 patients, including 12 healthy conjunctivas, 12 melanomas, seven squamous cell carcinomas, and seven papilloma samples, were analyzed using high-throughput RNA sequencing to assess messenger RNA (mRNA) expression of the SARS-CoV-2 receptor ACE2 and its cofactors including TMPRSS2, ANPEP, DPP4, and ENPEP. ACE2 protein expression was assessed in eight healthy conjunctival samples using immunohistochemistry. Our results show that the SARS-CoV-2 receptor ACE2 is not substantially expressed in conjunctival samples on the mRNA (median: 0.0 transcripts per million [TPM], min: 0.0 TPM, max: 1.7 TPM) and protein levels. Similar results were obtained for the transcription of other auxiliary molecules. In conclusion, this study finds no evidence for a significant expression of ACE2 and its auxiliary mediators for cell entry in conjunctival samples, making conjunctival infection with SARS-CoV-2 via these mediators unlikely.


Assuntos
COVID-19/virologia , Carcinoma de Células Escamosas/virologia , Neoplasias Oculares/virologia , Melanoma/virologia , Papiloma/virologia , Receptores Virais/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/complicações , COVID-19/patologia , COVID-19/cirurgia , Carcinoma de Células Escamosas/complicações , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/cirurgia , Estudos de Casos e Controles , Túnica Conjuntiva/patologia , Túnica Conjuntiva/cirurgia , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Neoplasias Oculares/complicações , Neoplasias Oculares/patologia , Neoplasias Oculares/cirurgia , Expressão Gênica , Glutamil Aminopeptidase/genética , Glutamil Aminopeptidase/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Melanoma/complicações , Melanoma/patologia , Melanoma/cirurgia , Pessoa de Meia-Idade , Papiloma/complicações , Papiloma/patologia , Papiloma/cirurgia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Virais/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
6.
Int J Cancer ; 145(4): 974-978, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30694555

RESUMO

Talimogene laherparepvec (T-VEC) is a modified herpes simplex virus, type 1 (HSV-1), which can be administered intralesionally in patients with stage IIIB/C-IVM1a unresectable melanoma (EMA label). The phase 3 OPTiM registration study showed an overall response rate (ORR) of 26%. Since December 2016, 48 eligible patients started treatment at the Netherlands Cancer Institute. We included 26 patients in this study with a follow up time ≥6 months, reporting Overall Response Rate (ORR), Disease Control Rate (DCR), Adverse Events (AE), prior treatment for melanoma and baseline characteristics, documented in a prospectively maintained database. In house developed treatment protocol consists of clinical evaluation, periodic PET-CT and histological biopsies for response evaluation. Median follow-up was 12.5 months. Of 26 patients, 16 (61.5%) had a Complete Response (CR) as their best response. Seven (26.9%) patients had a Partial Response (PR) as their best response, 1 (3.8%) patient Stable Disease (SD) and 2 (7.7%) patients Progressive Disease (PD). Best ORR was 88.5%. DCR was 92.3%. Grade 1-2 AEs occurred in all patients. Mostly, these consisted of fatigue, influenza-like symptoms and injection site erythema. All patients underwent prior treatment. Prior treatment did not influence response or toxicity of T-VEC. Best ORR for T-VEC monotherapy at our institute was 88.5% with 61.5% achieving a CR. This prospective study for T-VEC in early metastatic (stage IIIB/C-IVM1a) melanoma demonstrated superior results to the phase 3 OPTiM study and confirms the role of oncolytic immunotherapy for melanoma.


Assuntos
Herpesvirus Humano 1/imunologia , Melanoma/imunologia , Melanoma/terapia , Melanoma/virologia , Metástase Neoplásica/imunologia , Metástase Neoplásica/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Imunoterapia/métodos , Injeções Intralesionais , Masculino , Pessoa de Meia-Idade , Países Baixos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Estudos Prospectivos , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/terapia , Neoplasias Cutâneas/virologia
7.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30045986

RESUMO

Herpes simplex virus 1 (HSV-1)-mediated oncolytic therapy is an emerging cancer treatment modality with potential effectiveness against a variety of malignancies. To better understand the interaction of HSV-1 with neoplastic cells, we inoculated three-dimensional (3D) cultures of human uveal melanoma cells with HSV-1. 3D melanoma cultures were established by placing tumor cells on the surface of a Matrigel matrix, which was followed by the growth of tumor cells on the matrix surface and invasion of the Matrigel matrix by some tumor cells to form multicellular tumor spheroids within the matrix. When established 3D melanoma cultures were inoculated with HSV-1 by placing virus on the surface of cultures, virus infection caused extensive death of melanoma cells growing on the surface of the 3D matrix and significantly decreased the number of tumor cell spheroids within the matrix. However, HSV-1 infection did not lead to a complete destruction of tumor cells in the 3D cultures during a 17-day observation period and, surprisingly, HSV-1 infection promoted the growth of some melanoma cells within the matrix as determined by the significantly increased size of residual viable multicellular tumor spheroids in virus-inoculated 3D cultures at 17 days after virus inoculation. Acyclovir treatment inhibited HSV-1-induced tumor cell killing but did not block the virus infection-induced increase in spheroid size. These findings suggest that although HSV-1 oncolytic virotherapy may cause extensive tumor cell killing, it may also be associated with the unintended promotion of the growth of some tumor cells.IMPORTANCE Cancer cells are exposed to HSV-1 during oncolytic virotherapy with the intention of killing tumor cells. Our observations reported here suggest that potential dangers of HSV-1 oncolytic therapy include promotion of growth of some tumor cells. Furthermore, our findings raise the possibility that HSV-1 infection of neoplastic cells during natural infections or vaccinations may promote the growth of tumors. Our study indicates that HSV-1 infection of 3D tumor cell cultures provides an experimental platform in which mechanisms of HSV-1-mediated promotion of tumor cell growth can be effectively studied.


Assuntos
Herpes Simples/complicações , Herpesvirus Humano 1/patogenicidade , Melanoma/patologia , Terapia Viral Oncolítica , Esferoides Celulares/patologia , Neoplasias Uveais/patologia , Replicação Viral , Proliferação de Células , Herpes Simples/virologia , Humanos , Melanoma/terapia , Melanoma/virologia , Esferoides Celulares/virologia , Células Tumorais Cultivadas , Neoplasias Uveais/terapia , Neoplasias Uveais/virologia
8.
J Surg Oncol ; 120(3): 558-564, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31264725

RESUMO

BACKGROUND AND OBJECTIVES: Patients with in-transit or limited cutaneous metastatic melanoma may benefit from intralesional injections with talimogene laherparepvec (TVEC), a modified oncolytic herpesvirus. However, its use in patients with adverse prognostic scores in a real-life clinical setting has not been studied. METHODS: We performed a two-center retrospective analysis of 40 patients with metastatic melanoma treated with TVEC from 2015-2017. Demographics, overall response, and survival after therapy were noted. RESULTS: Overall, there was a durable response rate of 40%; median progression-free survival (PFS) was 10.5 months and median overall survival (OS) was not reached. Bulky disease was associated with decreased OS (15.7 months vs not reached, P < .05) and mPFS (2.3 months vs not reached, P < .05), when compared with smaller tumors. Poor performance status (ECOG 2-3) was associated with worse OS (10.2 months vs not reached, P < .05) and PFS (2.1 months vs not reached, P < .05) compared to patients with ECOG 0-1. There was no difference in the outcomes with age greater than 75 or with prior therapies. Adverse events were relatively tolerable. CONCLUSIONS: These findings demonstrate that TVEC is an effective and safe treatment for metastatic melanoma in a real-life clinical setting, and suggest parameters to aid in appropriate therapy selection for optimal response.


Assuntos
Herpesviridae/fisiologia , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Neoplasias Cutâneas/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Feminino , Humanos , Masculino , Melanoma/virologia , Pessoa de Meia-Idade , Intervalo Livre de Progressão , Estudos Retrospectivos , Neoplasias Cutâneas/virologia , Taxa de Sobrevida
9.
Jpn J Clin Oncol ; 49(3): 201-209, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30462296

RESUMO

Oncolytic virus therapy is a promising new option for cancer. It utilizes genetically engineered or naturally occurring viruses that selectively replicate in and kill cancer cells without harming normal cells. T-VEC (talimogene laherparepvec), a second-generation oncolytic herpes simplex virus type 1, was approved by the US Food and Drug Administration for the treatment of inoperable melanoma in 2015 and subsequently approved in Europe in 2016. Other oncolytic viruses using different parental viruses have also been tested in Phase III clinical trials and are ready for drug approval: Pexa-Vec (pexastimogene devacirepvec), an oncolytic vaccinia virus, CG0070, an oncolytic adenovirus, and REOLYSIN (pelareorep), an oncolytic reovirus. In Japan, as of May 2018, several oncolytic viruses have been developed, and some have already proceeded to clinical trials. In this review, we summarize clinical trials assessing oncolytic virus therapy that were conducted or are currently ongoing in Japan, specifically, T-VEC, the abovementioned oncolytic herpes simplex virus type 1, G47Δ, a third-generation oncolytic herpes simplex virus type 1, HF10, a naturally attenuated oncolytic herpes simplex virus type 1, Telomelysin, an oncolytic adenovirus, Surv.m-CRA, another oncolytic adenovirus, and Sendai virus particle. In the near future, oncolytic virus therapy may become an important and major treatment option for cancer in Japan.


Assuntos
Melanoma/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Ensaios Clínicos como Assunto , Humanos , Japão , Melanoma/virologia
10.
J Cell Physiol ; 233(12): 9052-9059, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29968911

RESUMO

The uveal melanoma (UM) is the most common human intraocular tumor. The BK polyomavirus (BKPyV) is a small DNA tumor virus whose footprints have been detected in different human cancers. BKPyV has oncogenic potential. Indeed, BKPyV, when inoculated into experimental animals, induces tumors of different histotypes, whereas in vitro, it transforms mammalian cells, including human cells from distinct tissues. In this investigation, the association between UM and BKPyV was studied employing indirect enzyme-linked immunosorbent assays (ELISAs) using synthetic peptides that mimic BKPyV viral capsid 1 (VP1) antigens. Indirect ELISAs were used to detect serum IgG antibodies against this polyomavirus with oncogenic potential in samples from patients with UM and controls, represented by healthy subjects (HS). It was found that serum samples from patients with UM had a higher prevalence of BKPyV antibodies, 85% (51/60), compared with that detected in HS1, 62% (54/87), and HS2, 57% (68/120). The different prevalence of BKPyV antibodies detected in UM versus the two control groups, HS1 and HS2, is statistically significant (p < 0.005). Our immunologic data suggest a significantly higher prevalence of antibodies against BKPyV VP1 epitopes in serum samples from patients with UM compared with HS. These results indicate an association between UM and BKPyV, suggesting that this small DNA tumor virus may be a cofactor in the UM onset or progression.


Assuntos
Anticorpos/sangue , Vírus BK/isolamento & purificação , Imunoglobulina G/sangue , Melanoma/sangue , Neoplasias Uveais/sangue , Idoso , Anticorpos/imunologia , Vírus BK/imunologia , Vírus BK/patogenicidade , Carcinogênese/genética , Carcinogênese/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoglobulina G/imunologia , Masculino , Melanoma/imunologia , Melanoma/virologia , Pessoa de Meia-Idade , Infecções por Polyomavirus/sangue , Infecções por Polyomavirus/imunologia , Infecções por Polyomavirus/virologia , Infecções Tumorais por Vírus/sangue , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia , Neoplasias Uveais/imunologia , Neoplasias Uveais/virologia
12.
Mol Biol (Mosk) ; 52(4): 659-666, 2018.
Artigo em Russo | MEDLINE | ID: mdl-30113031

RESUMO

The oncolytic potential of the attenuated mumps virus (MV) vaccine strain Leningrad-3 (L-3) was evaluated in a panel of four human metastatic melanoma cell lines. The lines were shown to be susceptible and permissive to MV infection. Efficient MV replication led to death of melanoma cells, but the effect differed among the cell lines. Possible mechanisms mediating the selectivity of MV L-3 towards the cell lines were explored. Replicative and oncolytic activity of MV was found to depend on the expression pattern of type I interferon genes. None of the melanoma cell lines showed induction of expression of the total spectrum of genes required to inhibit virus replication. Based on the results, MV L-3 was assumed to be a promising oncolytic agent for human melanoma cells.


Assuntos
Melanoma/terapia , Vírus da Caxumba/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/patologia , Melanoma/virologia , Camundongos , Proteínas de Neoplasias/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Curr Opin Oncol ; 29(5): 359-365, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28678032

RESUMO

PURPOSE OF REVIEW: The present review will highlight recent advances in the clinical application of oncolytic viral therapy. RECENT FINDINGS: Until recently, oncolytic viral researchers saw the immune system as an enemy that would clear the virus from the bloodstream. However, researchers now understand that sustained responses are seen in those patients with more robust antitumor immune responses. Much of the current focus in oncolytic viral research is trained on manipulation of the immune system to affect cancer cell killing in the tumor microenvironment and to facilitate durable systemic antitumor immunity. Many investigators have demonstrated synergistic effects of checkpoint inhibition and other immune therapies with viral administration. At the same time, insertion of various markers enables noninvasive deep tissue imaging. Finally, following regulatory approval in the United States and Europe, unbridled clinical use of T-VEC for patients with metastatic melanoma is also generating large volumes of patient data that will help elucidate strengths and weaknesses of oncolytic viral therapy. Perhaps the most telling sign of the field's future is a seismic shift in clinical trials with more investigators combining virus and immunotherapies. SUMMARY: This article reviews the current state of therapeutic oncolytic viruses in clinical use, and explores future directions of the field.


Assuntos
Melanoma/terapia , Terapia Viral Oncolítica/métodos , Ensaios Clínicos como Assunto , Humanos , Melanoma/imunologia , Melanoma/virologia , Terapia Viral Oncolítica/tendências , Vírus Oncolíticos
14.
Microbiol Immunol ; 61(11): 497-506, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28940687

RESUMO

Isolation of human metapneumovirus (HMPV) from clinical specimens is currently inefficient because of the lack of a cell culture system in which a distinct cytopathic effect (CPE) occurs. The cell lines LLC-MK2, Vero and Vero E6 are used for isolation of HMPV; however, the CPE in these cell lines is subtle and usually requires a long observation period and sometimes blind passages. Thus, a cell line in which an early and distinct CPE occurs following HMPV inoculation is highly desired by clinical virology laboratories. In this study, it was demonstrated that, in the human malignant melanoma cell line MNT-1, obvious syncytium formation occurs shortly after inoculation with HMPV-positive clinical specimens. In addition, the growth and efficiency of isolation of HMPV were greater using MNT-1 than using any other conventional cell line. Addition of this cell line to our routine viral isolation system for clinical specimens markedly enhanced isolation frequency, allowing isolation-based surveillance. MNT-1 has the potential to facilitate clinical and epidemiological studies of HMPV.


Assuntos
Melanoma/virologia , Metapneumovirus/fisiologia , Neoplasias Cutâneas/virologia , Linhagem Celular Tumoral , Efeito Citopatogênico Viral , Humanos , Metapneumovirus/genética , Metapneumovirus/crescimento & desenvolvimento , Metapneumovirus/isolamento & purificação , Melanoma Maligno Cutâneo
15.
Virus Genes ; 53(3): 477-482, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28188458

RESUMO

Oncolytic viruses (OVs) have emerged as a promising approach for melanoma treatment by causing tumor lysis and inducing immuno-modulatory activities. Tanapoxvirus (TPV), which causes a mild self-limiting disease in humans and contains a large DNA genome, appears as a promising OV candidate. TPV recombinants were generated with the thymidine kinase/66R gene deletion (TPVΔ66R), the 15L gene deletion (TPVΔ15L), or with both the 15L and 66R gene ablation (TPVΔ15LΔ66R). Our previous studies have shown that treatment of TPVΔ15L resulted in significant tumor regression in xenotransplanted human melanoma in nude mice. Here, we demonstrate that an anti-viral activity identified as interferon-λ1 (IFN-λ1) was secreted in a remarkably higher quantity from human lung fibroblast WI-38 and melanoma SK-MEL-3 cells infected with TPVΔ15L. Furthermore, we show that IFN-λ1 exhibits a more pronounced anti-proliferative effect in melanoma cells than IFN-α and IFN-ß in vitro. Additional experiments strongly suggest that TPVΔ15L kills melanoma cells partially through inducing IFN-λ1. Taken together, our results demonstrate the immuno-modulatory activities associated with TPVΔ15L and suggest further exploration of TPVΔ15L as a melanoma virotherapy.


Assuntos
Proliferação de Células , Citocinas/metabolismo , Melanoma/terapia , Melanoma/virologia , Proteínas Virais/metabolismo , Yatapoxvirus/genética , Animais , Antivirais/farmacologia , Linhagem Celular Tumoral , Fibroblastos/imunologia , Fibroblastos/virologia , Deleção de Genes , Vetores Genéticos , Interferon-alfa , Interferon beta , Pulmão/virologia , Melanoma/imunologia , Melanoma/patologia , Camundongos , Camundongos Nus , Neurregulinas/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Deleção de Sequência , Timidina Quinase/genética , Proteínas Virais/genética , Replicação Viral
16.
Ann Pharmacother ; 51(8): 675-681, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28351167

RESUMO

OBJECTIVE: To review the efficacy and safety of talimogene laherparepvec (T-VEC) as well as its pharmacology, pharmacokinetics, drug-drug interactions, handling procedures, cost considerations, and place in therapy. DATA SOURCES: Searches of PubMed (1966 to February 2017) and Cochrane Library (1999 to February 2017) were conducted using the terms talimogene laherparepvec, T-VEC, OncoVEX, immunotherapy, melanoma, and oncolytic virus. Additional information was determined from bibliographies, manufacturer product labeling and website, meeting abstracts, Food and Drug Administration website, and clinicaltrials.gov. STUDY SELECTION AND DATA EXTRACTION: A total of 79 English-language publications were identified. Articles that assessed T-VEC's pharmacokinetics, pharmacodynamics, mechanism, dosing, safety, and efficacy were included as well as narrative reviews that provided practical information. DATA SYNTHESIS: Clinical trials have confirmed the safety and efficacy of T-VEC as monotherapy for the treatment of advanced melanoma, with an overall response rate (ORR) of 26%. Relative to granulocyte-macrophage colony-stimulating factor, T-VEC significantly increased durable response rate (DRR; 16.3% vs 2.1%, P < 0.001); however, median overall survival was not improved (23.3 vs 18.9 months, P = 0.051). Phase 1b trials have combined T-VEC and immunotherapies with promising results. T-VEC's adverse effects are generally considered mild to moderate in severity. CONCLUSION: T-VEC is the first approved oncolytic virus for local treatment of unresectable cutaneous, subcutaneous, and nodal lesions in melanoma recurrent after initial surgery. T-VEC improves ORR and DRR as a single agent, shows promise in combination therapy, and is well tolerated. Ongoing trials will determine if T-VEC has a role in early treatment or in combination therapy for melanoma or other malignancies.


Assuntos
Imunoterapia/métodos , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Administração Cutânea , Ensaios Clínicos como Assunto , Terapia Combinada , Intervalo Livre de Doença , Interações Medicamentosas , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Humanos , Melanoma/patologia , Melanoma/virologia , Vírus Oncolíticos/genética
17.
Jpn J Clin Oncol ; 47(2): 175-178, 2017 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-28173241

RESUMO

After 4 weeks of the last dose of nivolumab, a 59-year-old man with stage IV melanoma was subject to treatment with ipilimumab. After 5 weeks, the patient developed severe hepatitis, showing markedly elevated levels of both aspartate aminotransferase and alanine aminotransferase (>2000 U/l). Using pulse steroid therapy with 1000 mg/d of methylprednisolone, liver function initially improved, but then deteriorated upon dosage reduction. Subsequently, mycophenolate mofetil (MMF) was administered at a dose of 2 g/d in addition to the corticosteroid, which resulted in aspartate aminotransferase and alanine aminotransferase levels gradually improving to grade 1, and the corticosteroid dose was successfully reduced to 0.5 mg/kg/d of oral prednisolone. Liver function then remained stable when MMF was tapered. In conclusion, the use of MMF improved liver function in this patient with steroid-refractory hepatitis induced by immune checkpoint inhibitor administration.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Ipilimumab/efeitos adversos , Melanoma/virologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Humanos , Ipilimumab/uso terapêutico , Masculino , Melanoma/tratamento farmacológico , Pessoa de Meia-Idade , Ácido Micofenólico/uso terapêutico , Nivolumabe
18.
Microbiol Immunol ; 60(11): 801-805, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27762443

RESUMO

Isolation of human parainfluenza virus (HPIV) serotypes 1 and 3 from clinical specimens is not very efficient because of the lack of a cell culture system capable of inducing CPE. In this study, the utility of a melanoma cell line, MNT-1, that allows HPIV growth and displays CPE was demonstrated. In particularly, the efficiency of isolating HPIV1 and HPIV3 using MNT-1 was greater than for cell lines conventionally used for HPIV isolation. Our demonstrated efficacy of HPIV1 and HPIV3 isolation with apparent CPE using the MNT-1 cell culture system has the potential to improve virus isolation from clinical specimens.


Assuntos
Vírus da Parainfluenza 1 Humana/isolamento & purificação , Vírus da Parainfluenza 1 Humana/fisiologia , Vírus da Parainfluenza 3 Humana/isolamento & purificação , Vírus da Parainfluenza 3 Humana/fisiologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Células Cultivadas , Efeito Citopatogênico Viral , Humanos , Melanoma/virologia , Infecções por Respirovirus/virologia
19.
Proc Natl Acad Sci U S A ; 110(5): 1911-6, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23322733

RESUMO

Herpesvirus entry functions of the conserved glycoproteins gB and gH-gL have been delineated, but their role in regulating cell-cell fusion is poorly understood. Varicella-zoster virus (VZV) infection provides a valuable model for investigating cell-cell fusion because of the importance of this process for pathogenesis in human skin and sensory ganglia. The present study identifies a canonical immunoreceptor tyrosine-based inhibition motif (ITIM) in the gB cytoplasmic domain (gBcyt) and demonstrates that the gBcyt is a tyrosine kinase substrate. Orbitrap mass spectrometry confirmed that Y881, central to the ITIM, is phosphorylated. To determine whether the gBcyt ITIM regulates gB/gH-gL-induced cell-cell fusion in vitro, tyrosine residues Y881 and Y920 in the gBcyt were substituted with phenylalanine separately or together. Recombinant viruses with these substitutions were generated to establish their effects on syncytia formation in replication in vitro and in the human skin xenograft model of VZV pathogenesis. The Y881F substitution caused significantly increased cell-cell fusion despite reduced cell-surface gB. Importantly, the Y881F or Y881/920F substitutions in VZV caused aggressive syncytia formation, reducing cell-cell spread. These in vitro effects of aggressive syncytia formation translated to severely impaired skin infection in vivo. In contrast, the Y920F substitution did not affect virus replication in vitro or in vivo. These observations suggest that gB modulates cell-cell fusion via an ITIM-mediated Y881 phosphorylation-dependent mechanism, supporting a unique concept that intracellular signaling through this gBcyt motif regulates VZV syncytia formation and is essential for skin pathogenesis.


Assuntos
Herpesvirus Humano 3/metabolismo , Motivo de Inibição do Imunorreceptor Baseado em Tirosina , Pele/patologia , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Células CHO , Fusão Celular , Linhagem Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Células Gigantes/ultraestrutura , Células Gigantes/virologia , Células HEK293 , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiologia , Humanos , Melanoma/patologia , Melanoma/ultraestrutura , Melanoma/virologia , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Mutação , Fosforilação , Estrutura Terciária de Proteína , Pele/virologia , Transplante Heterólogo , Tirosina/genética , Tirosina/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
20.
Vestn Ross Akad Med Nauk ; (2): 121-7, 2016.
Artigo em Russo | MEDLINE | ID: mdl-27522713

RESUMO

Melanoma is one of the most aggressive human malignant tumors. Its incidence and mortality are growing steadily. Ultraviolet irradiation is the main risk factor for melanoma involved in melanomagenesis. The probability of viral etiology of melanoma has been discussed. Human papillomaviruses (HPV) have been mentioned among candidates for its etiologic agents because some HPV types are the powerful carcinogens causing cervical cancer and other cancers. The review analyses the literature data on the association of melanoma with HPV Several groupsfound HPVin skin melanomas as well as in mucosa; viruses of high oncogenic risk were detected in some cases. For some organs the etiological role of high-risk HPV as inducers of invasive carcinomas is confirmed. These organs require special mention: cervix uteri, vulva, vagina, penis, anal region, and oral cavity. However in the majority of the studies in which viral DNA-positive melanomas were found, testing for viral genome expression was not done while this is the fact of primary importance. HPVare found in normal skin and mucous membranes thus creating justifiable threat of tumor specimen contamination with viral DNA in vivo. There are limited data on aggravation of the disease prognosis in papillomavirus-positive melanomas. However, any systematic observation of a sizeable patient group distinguished by that tumor type has not been performed yet. Viral E6 and E7 oncogenes of high-risk papillomaviruses were shown to be able to transform normal human melanocytes in vitro experiments. Thus, we can assume the presence of the association of melanoma with oncogenic HPV. The clinical significance of this problem is indisputable under the conditions of the steady increase in melanoma incidence and mortality rates in Russia and abroad. The problem requires further study.


Assuntos
Melanoma , Papillomaviridae , DNA Viral/análise , Humanos , Melanoma/etiologia , Melanoma/virologia , Papillomaviridae/isolamento & purificação , Papillomaviridae/fisiologia , Proteínas E7 de Papillomavirus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA