Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(6): e0037223, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37199666

RESUMO

Viral oncolytic immunotherapy is a nascent field that is developing tools to direct the immune system to find and eliminate cancer cells. Safety is improved by using cancer-targeted viruses that infect or grow poorly on normal cells. The recent discovery of the low-density lipoprotein (LDL) receptor as the major vesicular stomatitis virus (VSV) binding site allowed for the creation of a Her2/neu-targeted replicating recombinant VSV (rrVSV-G) by eliminating the LDL receptor binding site in the VSV-G glycoprotein (gp) and adding a sequence coding for a single chain antibody (SCA) to the Her2/neu receptor. The virus was adapted by serial passage on Her2/neu-expressing cancer cells resulting in a virus that yielded a 15- to 25-fold higher titer following in vitro infection of Her2/neu+-expressing cell lines than that of Her2/neu-negative cells (~1 × 108/mL versus 4 × 106 to 8 × 106/mL). An essential mutation resulting in a higher titer virus was a threonine-to-arginine change that produced an N-glycosylation site in the SCA. Infection of Her2/neu+ subcutaneous tumors yielded >10-fold more virus on days 1 and 2 than Her2/neu- tumors, and virus production continued for 5 days in Her2/neu+ tumors compared with 3 days that of 3 days in Her2/neu- tumors. rrVSV-G cured 70% of large 5-day peritoneal tumors compared with a 10% cure by a previously targeted rrVSV with a modified Sindbis gp. rrVSV-G also cured 33% of very large 7-day tumors. rrVSV-G is a new targeted oncolytic virus that has potent antitumor capabilities and allows for heterologous combination with other targeted oncolytic viruses. IMPORTANCE A new form of vesicular stomatitis virus (VSV) was created that specifically targets and destroys cancer cells that express the Her2/neu receptor. This receptor is commonly found in human breast cancer and is associated with a poor prognosis. In laboratory tests using mouse models, the virus was highly effective at eliminating implanted tumors and creating a strong immune response against cancer. VSV has many advantages as a cancer treatment, including high levels of safety and efficacy and the ability to be combined with other oncolytic viruses to enhance treatment results or to create an effective cancer vaccine. This new virus can also be easily modified to target other cancer cell surface molecules and to add immune-modifying genes. Overall, this new VSV is a promising candidate for further development as an immune-based cancer therapy.


Assuntos
Neoplasias da Mama , Glicoproteínas , Terapia Viral Oncolítica , Vírus Oncolíticos , Vesiculovirus , Animais , Feminino , Humanos , Camundongos , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Glicoproteínas/genética , Glicoproteínas/metabolismo , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Vesiculovirus/genética , Vesiculovirus/metabolismo , Replicação Viral , Análise de Sobrevida
2.
Front Immunol ; 14: 1137054, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37033929

RESUMO

Simple, reliable methods to detect anti-tumor memory T-cells are necessary to develop a clinical tumor vaccination program. A mouse model of curative viral onco-immunotherapy found that peritoneal tumor challenge following cure identified an oligoclonal anti-tumor memory CD4 and CD8 T-cell response. Clonotypes differed among the challenged animals but were congruent in blood, spleen and peritoneal cells (PC) of the same animal. Adoptive transfer demonstrated that the high-frequency responding T-cells were tumor specific. Tetramer analysis confirmed that clonotype frequency determined by T-cell receptor (TCR)- chain (TRB) analysis closely approximated cell clone frequency. The mean frequency of resting anti-tumor memory CD4 T-cells in unchallenged spleen was 0.028% and of memory CD8 T-cells was 0.11% which was not high enough to distinguish them from background. Stimulation produced a mean ~10-fold increase in splenic and 100-fold increase in peritoneal anti-tumor memory T-cell clonotypes. This methodology can be developed to use blood and tissue sampling to rapidly quantify the effectiveness of a tumor vaccine or any vaccine generating therapeutic T-cells.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Camundongos , Animais , Receptores de Antígenos de Linfócitos T , Neoplasias/terapia , Células Clonais , Transferência Adotiva
3.
J Child Neurol ; 37(1): 73-79, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34816755

RESUMO

Introduction: Continuous neurologic assessment in the pediatric intensive care unit is challenging. Current electroencephalography (EEG) guidelines support monitoring status epilepticus, vasospasm detection, and cardiac arrest prognostication, but the scope of brain dysfunction in critically ill patients is larger. We explore quantitative EEG in pediatric intensive care unit patients with neurologic emergencies to identify quantitative EEG changes preceding clinical detection. Methods: From 2017 to 2020, we identified pediatric intensive care unit patients at a single quaternary children's hospital with EEG recording near or during acute neurologic deterioration. Quantitative EEG analysis was performed using Persyst P14 (Persyst Development Corporation). Included features were fast Fourier transform, asymmetry, and rhythmicity spectrograms, "from-baseline" patient-specific versions of the above features, and quantitative suppression ratio. Timing of quantitative EEG changes was determined by expert review and prespecified quantitative EEG alert thresholds. Clinical detection of neurologic deterioration was defined pre hoc and determined through electronic medical record documentation of examination change or intervention. Results: Ten patients were identified, age 23 months to 27 years, and 50% were female. Of 10 patients, 6 died, 1 had new morbidity, and 3 had good recovery; the most common cause of death was cerebral edema and herniation. The fastest changes were on "from-baseline" fast Fourier transform spectrograms, whereas persistent changes on asymmetry spectrograms and suppression ratio were most associated with morbidity and mortality. Median time from first quantitative EEG change to clinical detection was 332 minutes (interquartile range: 201-456 minutes). Conclusion: Quantitative EEG is potentially useful in earlier detection of neurologic deterioration in critically ill pediatric intensive care unit patients. Further work is required to quantify the predictive value, measure improvement in outcome, and automate the process.


Assuntos
Cuidados Críticos/métodos , Eletroencefalografia/métodos , Unidades de Terapia Intensiva Pediátrica , Doenças do Sistema Nervoso/diagnóstico , Doença Aguda , Adolescente , Adulto , Criança , Pré-Escolar , Estado Terminal , Estudos de Avaliação como Assunto , Feminino , Humanos , Lactente , Masculino , Valor Preditivo dos Testes , Adulto Jovem
4.
Anticancer Res ; 38(12): 6621-6629, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30504370

RESUMO

BACKGROUND: Late developing breast cancer metastases are common and lethal despite treatment with adjuvant chemotherapy at the time of primary tumor excision. Stimulation of an antitumor immune response is an alternative strategy for preventing this devastating development. MATERIALS AND METHODS: A mouse model of the human epidermal growth factor receptor 2 (HER2/neu)-positive mammary cancer was used to compare the antitumor memory T-cell response following tumor cure by viral oncolytic immunotherapy, chemotherapy, surgical excision, or surgical excision plus virus infection. Memory T-cell response was assessed by functional in vivo assays. RESULTS: Antitumor T-cell memory was generated most powerfully by curative viral oncolytic immunotherapy and poorly by curative chemotherapy. Cure by surgical excision generated an immune antitumor response which was increased by neo-adjuvant virus infection. CD4 memory T-cells were most potent. CONCLUSION: Virus infection of tumor generates an antitumor memory immune response and chemotherapy suppresses this response. Clinical trials testing adjuvant immune stimulation instead of chemotherapy may be worth exploring because memory antitumor T-cells have the unique potential to find and eliminate small nests of metastatic cancer cells in sanctuary sites and prevent emergence of tumors from dormant cancer cells.


Assuntos
Antineoplásicos/uso terapêutico , Linfócitos T CD4-Positivos/fisiologia , Memória Imunológica/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Imunoterapia/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Terapia Viral Oncolítica/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Anticancer Res ; 38(1): 147-157, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29277767

RESUMO

BACKGROUND: Late metastases develop from cancer of the breast, prostate, lung, kidney and malignant melanomas. Memory T-cells have excellent potential to prevent this devastating development in the same way that they routinely prevent emergence of latent viruses. MATERIAL AND METHODS: A peritoneal tumor mouse model of viral oncotherapy was used to generate therapeutic antitumor memory T-cells. Functional in vivo and in vitro assays were used to study the temporal evolution of their anticancer effects. RESULTS: Highly therapeutic antitumor memory was generated by viral oncolytic immunotherapy 30 days after treatment and matured to maximal potency at 100 days. Maturation was not uniform across different measures. CONCLUSION: The results provide guidelines for developing a viral oncolytic vaccine strategy to generate antitumor memory T-cells that can eliminate small nests of metastatic cancer cells in sanctuary sites and prevent emergence of tumors from dormant cancer cell collections. The results are relevant to any immunization strategy designed to generate antitumor memory T-cells.


Assuntos
Imunoterapia , Terapia Viral Oncolítica , Neoplasias Peritoneais/terapia , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Diferenciação Celular , Modelos Animais de Doenças , Feminino , Camundongos Endogâmicos BALB C , Neoplasias Peritoneais/imunologia , Vesiculovirus
6.
Anticancer Res ; 35(9): 4593-7, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26254347

RESUMO

Cancer immune therapy is difficult partly because several classes of suppressor cells, including regulatory T-cells and macrophage-derived suppressor cells, inhibit the antitumor T-cell response. We used treatment studies of implanted tumors in mice to demonstrate that the same inhibitory cells that abrogated an acute therapeutic T-cell response to established tumor did not inhibit the therapeutic response produced by memory T-cells. Generating antitumor memory T-cells may be a highly potent strategy against cancer with late developing metastases.


Assuntos
Memória Imunológica , Neoplasias/imunologia , Linfócitos T/imunologia , Animais , Ciclofosfamida/farmacologia , Feminino , Humanos , Memória Imunológica/efeitos dos fármacos , Imunoterapia , Camundongos Endogâmicos BALB C , Neoplasias/patologia , Recombinação Genética/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
7.
Neuro Oncol ; 17(4): 536-44, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25223975

RESUMO

BACKGROUND: Leptomeningeal metastases occur in 2%-5% of patients with breast cancer and have an exceptionally poor prognosis. The blood-brain and blood-meningeal barriers severely inhibit successful chemotherapy. We have developed a straightforward method to induce antitumor memory T-cells using a Her2/neu targeted vesicular stomatitis virus. We sought to determine whether viral infection of meningeal tumor could attract antitumor memory T-cells to eradicate the tumors. METHODS: Meningeal implants in mice were studied using treatment trials and analyses of immune cells in the tumors. RESULTS: This paper demonstrates that there is a blood-meningeal barrier to bringing therapeutic memory T-cells to meningeal tumors. The barrier can be overcome by viral infection of the tumor. Viral infection of the meningeal tumors followed by memory T-cell transfer resulted in 89% cure of meningeal tumor in 2 different mouse strains. Viral infection produced increased infiltration and proliferation of transferred memory T-cells in the meningeal tumors. Following viral infection, the leukocyte infiltration in meninges and tumor shifted from predominantly macrophages to predominantly T-cells. Finally, this paper shows that successful viral therapy of peritoneal tumors generates memory CD8 T-cells that prevent establishment of tumor in the meninges of these same animals. CONCLUSIONS: These results support the hypothesis that a virally based immunization strategy can be used to both prevent and treat meningeal metastases. The meningeal barriers to cancer therapy may be much more permeable to treatment based on cells than treatment based on drugs or molecules.


Assuntos
Imunoterapia Adotiva , Neoplasias Meníngeas/terapia , Neoplasias Meníngeas/virologia , Linfócitos T/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Estimativa de Kaplan-Meier , Neoplasias Meníngeas/secundário , Camundongos , Receptor ErbB-2/metabolismo , Resultado do Tratamento , Vesiculovirus
8.
Int J Cancer ; 121(2): 425-30, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17354238

RESUMO

Vesicular stomatitis virus (VSV) is being developed for cancer therapy. We have created a recombinant replicating VSV (rrVSV) that targeted to Her2/neu expressing breast cancer cells and expresses mouse GM-CSF. We now tested the efficacy of this rrVSV in the treatment of peritoneal tumor implants of D2F2/E2 cells, a BALB/c mouse mammary tumor cell line, which was stably transfected to express Her2/neu. Mice were treated 1 day following tumor implantation with either 2 x 10(8) infectious doses rrVSV or conditioned media (CM). All control animals developed massive peritoneal tumor with a median survival of 16 days. Nine of 10 rrVSV treated mice survived long term with no evidence of tumor. rrVSV had much less efficacy in treating implants of the parent D2F2 cells that did not express Her2/neu. The median survival was 13.5 days in mice treated with CM and 21 days in those treated with rrVSV. There was one long term survivor in the rrVSV treated group. None of the rrVSV treated animals showed evidence of viral toxicity. Three of 7 long term survivors did not develop tumor when rechallenged first with D2F2/E2 and then with D2F2 cells. Both successful therapy and resistance to rechallenge were T-cell dependent. These studies demonstrate that targeted rrVSV eliminated peritoneal implants of Her2/neu expressing tumor and elicited an anti-tumor T-cell immunologic response.


Assuntos
Neoplasias Mamárias Experimentais/terapia , Receptor ErbB-2/metabolismo , Vírus da Estomatite Vesicular Indiana/genética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Feminino , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Interferon gama/metabolismo , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Terapia Viral Oncolítica/métodos , Receptor ErbB-2/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Análise de Sobrevida , Linfócitos T/citologia , Linfócitos T/imunologia , Fatores de Tempo , Resultado do Tratamento , Vírus da Estomatite Vesicular Indiana/fisiologia
9.
J Virol ; 80(17): 8603-12, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16912309

RESUMO

Vesicular stomatitis virus (VSV) is being developed for cancer therapy. We created a recombinant replicating VSV (rrVSV) that preferentially infected Her2/neu-expressing breast cancer cells. This rrVSV did not express the native VSV-G glycoprotein (gp). Instead, it expressed a chimeric Sindbis gp which included a single-chain antibody (SCA) directed to the human Her2/neu receptor. The virus infected mouse mammary carcinoma cells (D2F2/E2) expressing Her2/neu 23-fold better than the parent cells (D2F2). However, viral growth in cultured D2F2/E2 cells was curtailed after several cycles, and viral yield was very poor at 2 x 10(4) infectious doses (ID)/ml. We performed in vitro serial passage in D2F2/E2 cells to evolve a virus with improved growth that could be used for preclinical therapy trials in mice. Fifteen passes generated an adapted virus that progressed through multiple cycles in cultured D2F2/E2 cells until all cells were infected and had a viral yield of 1 x 10(8) ID/ml. Sequencing of the entire viral genomes found only 2 mutations in the adapted virus. Both mutations occurred in the gp gene segment coding for the SCA. An additional N-glycosylation site was created by one of the mutations. The adapted virus showed higher density of gp on the viral envelope, improved infectivity, much greater stability, higher burst size, and decreased induction of cellular interferon. The specificity for cells expressing the Her2/neu receptor was unchanged. These studies demonstrate that serial passage can be used to rapidly evolve a VSV genome encoding an improved chimeric glycoprotein.


Assuntos
Adaptação Fisiológica , Neoplasias da Mama/virologia , Carcinoma/virologia , Recombinação Genética , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Cricetinae , Feminino , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Mutação , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Inoculações Seriadas , Fatores de Tempo , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Replicação Viral
10.
Virology ; 330(1): 24-33, 2004 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-15527831

RESUMO

Vesicular stomatitis virus (VSV) is a candidate for development for cancer therapy. We created a recombinant replicating VSV (rrVSV) with an altered surface protein that targeted preferentially to breast cancer cells. The rrVSV genome contained a single glycoprotein (gp) gene derived from Sindbis virus. This gene expressed a chimeric Sindbis E2 binding gp and the native Sindbis E1 fusion gp. The chimeric E2 binding gp, called Sindbis-SCA-erbb2, was modified to reduce its native binding function and to contain a single chain antibody (SCA) with specificity for the human epidermal growth factor receptor Her2/neu protein, erbb2. These viruses selectively infected, replicated in and killed cells expressing erbb2. The titer of rrVSV on SKBR3 cells, a human breast cancer cell line which highly expresses erbb2 was 3.1 x 10(7)/ml compared with a titer of 7.3 x 10(5)/ml on 143 cells, a human osteosarcoma cell line which does not express erbb2. The titer of rrVSV on D2F2/E2 cells, a mouse mammary cancer cell line stably transfected to express human erbb2 was 2.46 x 10(6)/ml compared with a titer of 5 x 10(4)/ml on the parent D2F2 cells which do not express erbb2. When titered on erbb2-negative cells, non-replicating pseudotype VSV coated with Sindbis-SCA-erbb2 had <3% the titer of pseudotype VSV coated with wild type Sindbis gp indicating that the chimeric Sindbis gp had severely impaired binding to the natural receptor. Analysis of the protein composition of the rrVSV found low expression of the modified Sindbis gp on the virus.


Assuntos
Neoplasias da Mama/virologia , Vírus da Estomatite Vesicular Indiana/isolamento & purificação , Linhagem Celular Tumoral , Feminino , Genoma Viral , Humanos , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral
11.
Clin Cancer Res ; 10(20): 6985-92, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15501978

RESUMO

Labeling of specific antibodies with bifunctional chelated Actinium-225 ((225)Ac; an alpha generator) allows the formation of new, highly potent and selective alpha-emitting anticancer drugs. We synthesized and evaluated a radioimmunoconjugate based on 3F8, an IgG(3) antibody that specifically binds to ganglioside GD2, which is overexpressed by many neuroectodermal tumors including neuroblastoma. The (225)Ac-1,4,7,10-tetra-azacylododecane (DOTA)-3F8 construct was evaluated for radiochemical purity and sterility, immunoreactivity, cytotoxicity in vitro, induction of apoptosis on GD2-positive cells, as well as for pharmacological biodistribution and metabolism of the (225)Ac generator and its daughters in a nude mouse xenograft model of neuroblastoma. The (225)Ac-3F8 showed an IC(50) of 3 Bq/ml (80 pCi/ml) on the neuroblastoma cell line, NMB7, in vitro. Apoptosis of these cells was not observed. Biodistribution in mice showed specific targeting of a subcutaneous tumor; there was redistribution of the (225)Ac daughter nuclides mainly from blood to kidneys and to small intestine. Toxicity was examined in cynomolgus monkeys. Monkeys injected with 1 to 3 doses of intrathecal (225)Ac-3F8 radioimmunoconjugate (80 to 150 kBq/kg total dose) did not show signs of toxicity based on blood chemistry, complete blood counts, or by clinical evaluations. Therapeutic efficacy of intrathecal (225)Ac-3F8 was studied in a nude rat xenograft model of meningeal carcinomatosis. The (225)Ac-3F8 treatment improved survival 2-fold from 16 to 34 days (P = 0.01). In conclusion, in vivo alpha generators targeted by 3F8 warrant additional study as a possible new approach to the treatment of carcinomatous meningitis.


Assuntos
Actínio/uso terapêutico , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Carcinoma/tratamento farmacológico , Carcinoma/patologia , Gangliosídeos/metabolismo , Imunoglobulina G/imunologia , Imunoglobulina G/uso terapêutico , Neoplasias Meníngeas/tratamento farmacológico , Neoplasias Meníngeas/patologia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Actínio/farmacocinética , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Murinos , Haplorrinos , Humanos , Imunoconjugados , Injeções Espinhais , Camundongos , Ratos , Ratos Nus , Análise de Sobrevida , Distribuição Tecidual , Transplante Heterólogo
12.
Virology ; 316(2): 337-47, 2003 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-14644615

RESUMO

Vesicular stomatitis virus (VSV) is a candidate for development for cancer therapy. It is an oncolytic virus that is safe in humans. Recombinant virus can be made directly from plasmid components. We attempted to create a virus that targeted specifically to breast cancer cells. Nonreplicating and replicating pseudotype VSV were created whose only surface glycoprotein (gp) was a Sindbis gp, called Sindbis-ZZ, modified to severely reduce its native binding function and to contain the Fc-binding domain of Staphylococcus aureus protein A. When titered on Her2/neu overexpressing SKBR3 human breast cancer cells, pseudotype VSV coated with Sindbis-ZZ had <1% the titer of pseudotype VSV coated with wild-type Sindbis gp. Titer was increased 50-fold when the Sindbis-ZZ pseudotype was conjugated with 4D5, a mouse monoclonal antibody directed against the Her2/neu receptor. Titers of antibody-conjugated virus were increased 36-fold on a second human breast cancer cell line, MCF7/H2, which expressed lower concentrations of Her2/neu receptor on the cell surface. At multiple concentrations of antibody, titers on SKBR3 cells were significantly greater when the virus was incubated with Herceptin, an antibody with a human Fc, than with 4D5, a mouse antibody, reflecting the known higher affinity of the protein A Fc-binding domain for human Fc. Analysis of the protein composition of the pseudotype VSV found low expression of the modified Sindbis gp on the virus accounting, in part, for a viral titer that did not exceed 1.2 x 10(5)/ml. This work demonstrates the ability to easily create, directly from plasmid components, an oncolytic replicating VSV with a restricted host cell range.


Assuntos
Anticorpos Monoclonais/genética , Neoplasias da Mama/terapia , Terapia Genética/métodos , Fragmentos Fc das Imunoglobulinas/genética , Receptor ErbB-2/antagonistas & inibidores , Proteínas Recombinantes de Fusão/genética , Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/genética , Sítios de Ligação , Linhagem Celular Tumoral , Feminino , Humanos , Concentração de Íons de Hidrogênio , Proteína Estafilocócica A/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...