Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Rev Clin Oncol ; 21(7): 489-500, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38760500

RESUMO

mRNA vaccines have been revolutionary in terms of their rapid development and prevention of SARS-CoV-2 infections during the COVID-19 pandemic, and this technology has considerable potential for application to the treatment of cancer. Compared with traditional cancer vaccines based on proteins or peptides, mRNA vaccines reconcile the needs for both personalization and commercialization in a manner that is unique to each patient but not beholden to their HLA haplotype. A further advantage of mRNA vaccines is the availability of engineering strategies to improve their stability while retaining immunogenicity, enabling the induction of complementary innate and adaptive immune responses. Thus far, no mRNA-based cancer vaccines have received regulatory approval, although several phase I-II trials have yielded promising results, including in historically poorly immunogenic tumours. Furthermore, many early phase trials testing a wide range of vaccine designs are currently ongoing. In this Review, we describe the advantages of cancer mRNA vaccines and advances in clinical trials using both cell-based and nanoparticle-based delivery methods, with discussions of future combinations and iterations that might optimize the activity of these agents.


Assuntos
COVID-19 , Vacinas Anticâncer , Neoplasias , Vacinas de mRNA , Humanos , Vacinas Anticâncer/uso terapêutico , Vacinas Anticâncer/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/prevenção & controle , Neoplasias/genética , COVID-19/prevenção & controle , COVID-19/imunologia , SARS-CoV-2/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico , RNA Mensageiro/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Ensaios Clínicos como Assunto
2.
Front Immunol ; 14: 1116034, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37575220

RESUMO

Background: Monocytes and monocyte-derived tumor infiltrating cells have been implicated in the immunosuppression and immune evasion associated with pancreatic adenocarcinoma (PDAC). Yet, precisely how monocytes in the periphery and tumor microenvironment in patients with intraductal papillary mucinous neoplasm (IPMN), a precursor lesion to PDAC, change during disease progression has not been defined. Here we functionally profiled the peripheral immune system and characterized the tumor microenvironment of patients with both IPMN and PDAC. We also tested if sera from patients with IPMN and PDAC functionally reprogram monocytes relative to that of healthy donors. Methods: Pancreatic tissue and peripheral blood were collected at the time of resection from 16 patients with IPMN and 32 patients with PDAC. Peripheral blood and pancreatic tissue/tumor were immunophenotyped using flow cytometry. Whole blood was plated and incubated with R848 (a TLR 7/8 agonist) or LPS (a TLR4 agonist) for 6 hours and TNF expression in monocytes was measured by flow cytometry to measure monocyte activation. To test if TLR sensitivity is determined by factors in patient sera, we preconditioned healthy donor monocytes in serum from PDAC (n=23), IPMN (n=15), or age-matched healthy donors (n=10) followed by in vitro stimulation with R848 or LPS and multiplex cytokine measurements in the supernatant. Results: TNF expression in R848-stimulated peripheral blood monocytes was higher in patients with low grade vs high grade IPMN (65% vs 32%, p = 0.03) and stage 1 vs stage 2/3 PDAC (58% vs 42%, p = 0.03), this was not observed after LPS stimulation. TLR activation correlated with increasing grade of dysplasia from low grade IPMN to high grade IPMN. Serum from patients with IPMN and PDAC recapitulated suppression of TNF induction after R848 stimulation in naïve, healthy donor monocytes. Conclusion: Peripheral blood monocyte TNF secretion inversely correlates with the degree of dysplasia in IPMN and cancer stage in PDAC, suggesting innate immune reprogramming as IPMNs progress to invasive disease. These effects are, at least in part, mediated by soluble mediators in sera.


Assuntos
Adenocarcinoma Mucinoso , Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Intraductais Pancreáticas , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/patologia , Monócitos/metabolismo , Adenocarcinoma/patologia , Carcinoma Ductal Pancreático/patologia , Lipopolissacarídeos , Hiperplasia/patologia , Microambiente Tumoral , Neoplasias Pancreáticas
3.
Nucleic Acid Ther ; 32(6): 449-456, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36346283

RESUMO

The success of mRNA vaccines against COVID-19 is nothing short of a medical revolution. Given its chemical lability the use of mRNA as a therapeutic has been counterintuitive and met with skepticism. The development of mRNA-based COVID-19 vaccines was the culmination of long and painstaking efforts by many investigators spanning over 30 years and culminating with the seminal studies of Kariko and Weissman. This review will describe one chapter in this saga, studies that have shown that mRNA can function as a therapeutic. It started with our seminal observation that dendritic cells (DCs) transfected with mRNA in vitro administered to mice inhibits tumor growth, and led to first-in-human clinical trials with mRNA vaccines in cancer patients. The clinical development of this patient-specific DCs-mRNA approach and use on a larger scale was hindered by the challenges associated with personalized cell therapies. Confirmed and extended by many investigators, these studies did serve as impetus and motivation that led scientists to persevere, eventually leading to the development of simple, broadly applicable, and highly effective protocols of directly injecting mRNA into patients, culminating in the COVID-19 mRNA vaccines.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Humanos , Animais , Camundongos , Vacinas contra COVID-19/genética , COVID-19/prevenção & controle , Vacinas de mRNA
4.
Front Immunol ; 13: 952220, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36052068

RESUMO

Introduction: B cells are key regulators of immune responses in melanoma. We aimed to explore differences in the histologic location and activation status of B cell follicles in sentinel lymph nodes (SLN) of melanoma patients. Methods: Flow cytometry was performed on fresh tumor draining lymph nodes (LN). Paraffin slides from a separate cohort underwent NanoString Digital Spatial Profiling (DSP)®. After staining with fluorescent markers for CD20 (B cells), CD3 (T cells), CD11c (antigen presenting cells) and a nuclear marker (tumor) was performed, regions of interest (ROI) were selected based on the location of B cell regions (B cell follicles). A panel of 68 proteins was then analyzed from the ROIs. Results: B cell percentage trended higher in patients with tumor in LN (n=3) compared to patients with nSLN (n=10) by flow cytometry. B cell regions from a separate cohort of patients with tumor in the (pSLN) (n=8) vs. no tumor (nSLN) (n=16) were examined with DSP. Within B cell regions of the SLN, patients with pSLN had significantly higher expression of multiple activation markers including Ki-67 compared to nSLN patients. Among 4 patients with pSLN, we noted variability in arrangement of B cell follicles which were either surrounding the tumor deposit or appeared to be infiltrating the tumor. The B cell follicle infiltrative pattern was associated with prolonged recurrence free survival. Conclusion: These data suggest a role for B cell follicles in coordinating effective adaptive immune responses in melanoma when low volume metastatic disease is present in tumor draining LN.


Assuntos
Melanoma , Neoplasias Cutâneas , Biologia , Humanos , Excisão de Linfonodo , Metástase Linfática , Melanoma/patologia , Biópsia de Linfonodo Sentinela , Neoplasias Cutâneas/patologia
5.
J Immunother Cancer ; 10(9)2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36175036

RESUMO

BACKGROUND: We previously reported results from a phase 1 study testing intratumoral recombinant poliovirus, lerapolturev, in 12 melanoma patients. All 12 patients received anti-PD-1 systemic therapy before lerapolturev, and 11 of these 12 patients also received anti-PD-1 after lerapolturev. In preclinical models lerapolturev induces intratumoral innate inflammation that engages antitumor T cells. In the current study, prelerapolturev and postlerapolturev tumor biopsies and blood were evaluated for biomarkers of response. METHODS: The following analyses were performed on tumor tissue (n=11): (1) flow cytometric assessment of immune cell density, (2) NanoString Digital Spatial profiling of protein and the transcriptome, and (3) bulk RNA sequencing. Immune cell phenotypes and responsiveness to in vitro stimulation, including in vitro lerapolturev challenge, were measured in peripheral blood (n=12). RESULTS: Three patients who received anti-PD-1 therapy within 30 days of lerapolturev have a current median progression-free survival (PFS) of 2.3 years and had higher CD8+T cell infiltrates in prelerapolturev tumor biopsies relative to that of 7 patients with median PFS of 1.6 months and lower CD8+T cell infiltrates in prelerapolturev tumor biopsies. In peripheral blood, four patients with PFS 2.3 years (including three that received anti-PD-1 therapy within 30 days before lerapolturev and had higher pretreatment tumor CD8+T cell infiltrates) had significantly higher effector memory (CD8+, CCR7-, CD45RA-) but lower CD8+PD-1+ and CD4+PD-1+ cells compared with eight patients with median PFS 1.6 months. In addition, pretreatment blood from the four patients with median PFS 2.3 years had more potent antiviral responses to in vitro lerapolturev challenge compared with eight patients with median PFS 1.6 months. CONCLUSION: An inflamed pretreatment tumor microenvironment, possibly induced by prior anti-PD-1 therapy and a proficient peripheral blood pretreatment innate immune response (antiviral/interferon signaling) to lerapolturev was associated with long term PFS after intratumoral lerapolturev in a small cohort of patients. These findings imply a link between intratumoral T cell inflammation and peripheral immune function. TRIAL REGISTRATION NUMBER: NCT03712358.


Assuntos
Melanoma , Microambiente Tumoral , Humanos , Inflamação , Interferons , Melanoma/tratamento farmacológico , Prognóstico , Receptores CCR7
6.
Mol Ther ; 29(5): 1772-1781, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33348055

RESUMO

Tumor cells release nucleic acid-containing proinflammatory complexes, termed nucleic acid-containing damage-associated molecular patterns (NA DAMPs), passively upon death and actively during stress. NA DAMPs activate pattern recognition receptors on cells in the tumor microenvironment leading to prolonged and intensified inflammation that potentiates metastasis. No strategy exists to control endogenous or therapy-induced inflammation in cancer patients. We discovered that the generation 3.0 polyamidoamine dendrimer (PAMAM-G3) scavenges NA DAMPs and mitigates their proinflammatory effects. In this study, we tested if the nucleic acid scavenger (NAS) PAMAM-G3 reduces lung metastasis in murine models of breast cancer. Our data indicate that PAMAM-G3 treatment decreases cell-free DNA levels and reduces lung metastasis in the experimental intravenous tumor-injection model and the postsurgical tumor-resection model of 4T1 breast cancer. Reduction in lung metastasis is associated with reduction in inflammatory immune cell subsets and proinflammatory cytokine levels in the tumor and the periphery. This study is the first example of NAS-mediated inhibition of metastasis to the lung. The study results provide a strong rationale for inclusion of NAS therapy in women with breast cancer undergoing standard-of-care surgery.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Dendrímeros/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Administração Intravenosa , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Ácidos Nucleicos Livres/efeitos dos fármacos , Citocinas/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Immunity ; 48(1): 147-160.e7, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29343435

RESUMO

Despite recent advances, many cancers remain refractory to available immunotherapeutic strategies. Emerging evidence indicates that the tolerization of local dendritic cells (DCs) within the tumor microenvironment promotes immune evasion. Here, we have described a mechanism by which melanomas establish a site of immune privilege via a paracrine Wnt5a-ß-catenin-peroxisome proliferator-activated receptor-γ (PPAR-γ) signaling pathway that drives fatty acid oxidation (FAO) in DCs by upregulating the expression of the carnitine palmitoyltransferase-1A (CPT1A) fatty acid transporter. This FAO shift increased the protoporphyrin IX prosthetic group of indoleamine 2,3-dioxgenase-1 (IDO) while suppressing interleukin(IL)-6 and IL-12 cytokine expression, culminating in enhanced IDO activity and the generation of regulatory T cells. We demonstrated that blockade of this pathway augmented anti-melanoma immunity, enhanced the activity of anti-PD-1 antibody immunotherapy, and suppressed disease progression in a transgenic melanoma model. This work implicates a role for tumor-mediated metabolic reprogramming of local DCs in immune evasion and immunotherapy resistance.


Assuntos
Células Dendríticas/metabolismo , Melanoma/imunologia , Proteína Wnt-5a/metabolismo , beta Catenina/metabolismo , Animais , Linhagem Celular , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Ácidos Graxos/metabolismo , Feminino , Citometria de Fluxo , Immunoblotting , Masculino , Melanoma/metabolismo , Camundongos , Camundongos Transgênicos , PPAR gama/metabolismo , Comunicação Parácrina/fisiologia , Reação em Cadeia da Polimerase , Transdução de Sinais/fisiologia
8.
Sci Transl Med ; 9(408)2017 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-28931654

RESUMO

Tumors thrive in an immunosuppressive microenvironment that impedes antitumor innate and adaptive immune responses. Thus, approaches that can overcome immunosuppression and engage antitumor immunity are needed. This study defines the adjuvant and cancer immunotherapy potential of the recombinant poliovirus/rhinovirus chimera PVSRIPO. PVSRIPO is currently in clinical trials against recurrent World Health Organization grade IV malignant glioma, a notoriously treatment-refractory cancer. Cytopathogenic infection of neoplastic cells releases the proteome and exposes pathogen- and damage-associated molecular patterns. At the same time, sublethal infection of antigen-presenting cells, such as dendritic cells and macrophages, yields potent, sustained type I interferon-dominant activation in an immunosuppressed microenvironment and promotes the development of tumor antigen-specific T cell responses in vitro and antitumor immunity in vivo. PVSRIPO's immune adjuvancy stimulates canonical innate anti-pathogen inflammatory responses within the tumor microenvironment that culminate in dendritic cell and T cell infiltration. Our findings provide mechanistic evidence that PVSRIPO functions as a potent intratumor immune adjuvant that generates tumor antigen-specific cytotoxic T lymphocyte responses.


Assuntos
Antígenos de Neoplasias/metabolismo , Células Dendríticas/imunologia , Imunoterapia , Poliovirus/genética , Recombinação Genética/genética , Linfócitos T Citotóxicos/imunologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Imunidade , Terapia de Imunossupressão , Interferons/metabolismo , Macrófagos/metabolismo , Melanoma/imunologia , Melanoma/patologia , Melanoma/terapia , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Vírus Oncolíticos/fisiologia , Proteoma/metabolismo , RNA de Cadeia Dupla/metabolismo , Rhinovirus/fisiologia
9.
Oncotarget ; 7(48): 79828-79841, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27806313

RESUMO

Intratumoral inoculation of viruses with tumor-selective cytotoxicity may induce cancer cell death and, thereby, shrink neoplastic lesions. It is unlikely, however, that viral tumor cell killing alone could produce meaningful, durable clinical responses, as clinically suitable 'oncolytic' viruses are severely attenuated and their spread and propagation are opposed by host immunity. Thus, a more propitious event in this context is the innate antiviral response to intratumoral virus administration, in particular for recruiting durable adaptive immune effector responses. It may represent a double-edged sword, as innate immune activation may eliminate infected tumor cells early, intercept viral spread and block any meaningful therapeutic response. The innate response to viral infection of tumors may be very different from that in non-malignant target tissues, owing to the unusual composition/tissue properties of tumor stroma. In this work, we report investigations of the innate immune response to the oncolytic poliovirus recombinant, PVSRIPO, in two mouse xenotransplantation models for breast and prostate cancer. Our observations indicate short-term virus persistence in infected tumors and virus recovery indicative of modest intratumoral propagation and persistence. Yet, a powerful innate inflammatory response coincided with chemokine induction and myeloid cell infiltration into tumors that was, interestingly, dominated by neutrophils. The combined effect of PVSRIPO tumor infection and the innate response it elicits was significant tumor regression in both models.


Assuntos
Imunidade Inata , Neoplasias Inflamatórias Mamárias/terapia , Terapia Viral Oncolítica/métodos , Poliovirus , Neoplasias da Próstata/terapia , Animais , Linhagem Celular Tumoral , Feminino , Células HeLa , Humanos , Inflamação/imunologia , Inflamação/virologia , Neoplasias Inflamatórias Mamárias/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Vírus Oncolíticos/genética , Poliovirus/genética , Poliovirus/imunologia , Neoplasias da Próstata/imunologia , Transgenes , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Discov Med ; 22(119): 67-72, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27585232

RESUMO

The concept that RNA has played a major role in the evolution of life stems from the "RNA World" hypothesis. This role of RNA was not immediately appreciated. Similarly, the scientific community has just recently begun to recognize the true potential of RNA as the drug of choice for gene therapy, cellular reprogramming and vaccination. While it is perhaps the most unstable of the three most commonly used biotherapeutics, the others being DNA and protein, the advantages that using RNA presents are now being realized at a high rate. The development of methods to protect it from degradation and deliver it in vivo has fueled more research into uses that were once considered heretical. In this age of enlightenment we are seeing significant investments in the 'RNA approach' both in academia and industry. Thus, along with developing RNA encoding antigens for vaccine development for cancer and infectious diseases, RNA is now used to program cells in vivo or ex vivo. In the following review we have chosen to highlight a few of the most recent studies that use RNA as a means to alter a disease state. These papers were chosen to indicate the breadth of research that is ongoing and hopefully to inspire others to consider new ways to treat cancer, infectious disease, or genetic disorders with RNA-based approaches.


Assuntos
RNA/metabolismo , Animais , Doenças Transmissíveis/genética , Doenças Transmissíveis/metabolismo , DNA/genética , DNA/metabolismo , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA/genética
11.
J Neurooncol ; 125(1): 65-74, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26311248

RESUMO

Generation of patient-derived, autologous dendritic cells (DCs) is a critical component of cancer immunotherapy with ex vivo-generated, tumor antigen-loaded DCs. An important factor in the ability to generate DCs is the potential impact of prior therapies on DC phenotype and function. We investigated the ability to generate DCs using cells harvested from pediatric patients with medulloblastoma for potential evaluation of DC-RNA based vaccination approach in this patient population. Cells harvested from medulloblastoma patient leukapheresis following induction chemotherapy and granulocyte colony stimulating factor mobilization were cryopreserved prior to use in DC generation. DCs were generated from the adherent CD14+ monocytes using standard procedures and analyzed for cell recovery, phenotype and function. To summarize, 4 out of 5 patients (80%) had sufficient monocyte recovery to permit DC generation, and we were able to generate DCs from 3 out of these 4 patient samples (75%). Overall, we successfully generated DCs that met phenotypic requisites for DC-based cancer therapy from 3 out of 5 (60%) patient samples and met both phenotypic and functional requisites from 2 out of 5 (40%) patient samples. This study highlights the potential to generate functional DCs for further clinical treatments from refractory patients that have been heavily pretreated with myelosuppressive chemotherapy. Here we demonstrate the utility of evaluating the effect of the currently employed standard-of-care therapies on the ex vivo generation of DCs for DC-based clinical studies in cancer patients.


Assuntos
Neoplasias Encefálicas/patologia , Células Dendríticas/fisiologia , Quimioterapia de Indução , Leucaférese , Meduloblastoma/patologia , Antígenos CD/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Separação Celular , Criança , Técnicas de Cocultura , Testes Imunológicos de Citotoxicidade , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/patologia , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Meduloblastoma/tratamento farmacológico , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Survivina , Transdução Genética , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo
12.
Sci Rep ; 4: 4632, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24755960

RESUMO

Emergence of drug-resistant strains of the pathogen Mycobacterium tuberculosis (Mtb) and the ineffectiveness of BCG in curtailing Mtb infection makes vaccine development for tuberculosis an important objective. Identifying immunogenic CD8+ T cell peptide epitopes is necessary for peptide-based vaccine strategies. We present a three-tiered strategy for identifying and validating immunogenic peptides: first, identify peptides that form stable complexes with class I MHC molecules; second, determine whether cytotoxic T lymphocytes (CTLs) raised against the whole protein antigen recognize and lyse target cells pulsed with peptides that passed step 1; third, determine whether peptides that passed step 2, when administered in vivo as a vaccine in HLA-A2 transgenic mice, elicit CTLs that lyse target cells expressing the whole protein antigen. Our innovative approach uses dendritic cells transfected with Mtb antigen-encoding mRNA to drive antigen expression. Using this strategy, we have identified five novel peptide epitopes from the Mtb proteins Apa, Mtb8.4 and Mtb19.


Assuntos
Células Dendríticas/imunologia , Mapeamento de Epitopos/métodos , Epitopos de Linfócito T/imunologia , Ensaios de Triagem em Larga Escala , Antígenos de Histocompatibilidade Classe I/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos de Bactérias/imunologia , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidade Classe I/química , Camundongos , Camundongos Transgênicos , Mycobacterium tuberculosis/genética , Peptídeos/imunologia , Ligação Proteica/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia
13.
Clin Cancer Res ; 20(10): 2684-94, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24658154

RESUMO

PURPOSE: Despite aggressive conventional therapy, glioblastoma (GBM) remains uniformly lethal. Immunotherapy, in which the immune system is harnessed to specifically attack malignant cells, offers a treatment option with less toxicity. The expression of cytomegalovirus (CMV) antigens in GBM presents a unique opportunity to target these viral proteins for tumor immunotherapy. Although the presence of CMV within malignant gliomas has been confirmed by several laboratories, its relevance as an immunologic target in GBM has yet to be established. The objective of this study was to explore whether T cells stimulated by CMV pp65 RNA-transfected dendritic cells (DC) target and eliminate autologous GBM tumor cells in an antigen-specific manner. EXPERIMENTAL DESIGN: T cells from patients with GBM were stimulated with autologous DCs pulsed with CMV pp65 RNA, and the function of the effector CMV pp65-specific T cells was measured. RESULTS: In this study, we demonstrate the ability to elicit CMV pp65-specific immune responses in vitro using RNA-pulsed autologous DCs generated from patients with newly diagnosed GBM. Importantly, CMV pp65-specific T cells lyse autologous, primary GBM tumor cells in an antigen-specific manner. Moreover, T cells expanded in vitro using DCs pulsed with total tumor RNA demonstrated a 10- to 20-fold expansion of CMV pp65-specific T cells as assessed by tetramer analysis and recognition and killing of CMV pp65-expressing target cells. CONCLUSION: These data collectively demonstrate that CMV-specific T cells can effectively target glioblastoma tumor cells for immunologic killing and support the rationale for the development of CMV-directed immunotherapy in patients with GBM.


Assuntos
Citotoxicidade Imunológica/imunologia , Fosfoproteínas/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas da Matriz Viral/imunologia , Western Blotting , Células Cultivadas , Técnicas de Cocultura , Citocinas/imunologia , Citocinas/metabolismo , Citomegalovirus/imunologia , Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Testes Imunológicos de Citotoxicidade/métodos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Citometria de Fluxo , Glioblastoma/imunologia , Glioblastoma/patologia , Glioblastoma/virologia , Humanos , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Viral/genética , RNA Viral/imunologia , Linfócitos T/imunologia , Células Tumorais Cultivadas , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Adulto Jovem
14.
Adv Healthc Mater ; 3(6): 837-42, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24339387

RESUMO

The use of a cell-based vaccine composed of autologous whole blood cells loaded with mRNA is described. Mice immunized with whole blood cells loaded with mRNA encoding antigen develop anti-tumor immunity comparable to DC-RNA immunization. This approach offers a simple and affordable alternative to RNA-based cellular therapy by circumventing complex, laborious and expensive ex vivo manipulations required for DC-based immunizations.


Assuntos
Células Sanguíneas/metabolismo , Vacinas Anticâncer/imunologia , RNA Mensageiro/metabolismo , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Células Sanguíneas/citologia , Células Sanguíneas/transplante , Vacinas Anticâncer/genética , Vacinas Anticâncer/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Modelos Animais de Doenças , Eletroporação , Feminino , Imunoterapia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Melanoma Experimental/mortalidade , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/química , Taxa de Sobrevida
15.
J Clin Invest ; 123(7): 3135-45, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23934126

RESUMO

BACKGROUND: Many cancers, including melanoma, exclusively express constitutive proteasomes (cPs) and are unable to express immunoproteasomes (iPs). In contrast, mature DCs used for immunotherapy exclusively express iPs. Since proteasomes generate peptides presented by HLA class I molecules, we hypothesized that mature melanoma antigen-loaded DCs engineered to process antigens through cPs would be superior inducers of antimelanoma immunity in vivo. METHODS: Subjects with metastatic melanoma were vaccinated with mature DCs transfected with RNAs encoding melanoma antigens MART1, MAGE-3, gp100, and tyrosinase. These DCs were derived from monocytes that were untransfected (Arm A; n = 4), transfected with control siRNA (Arm B; n = 3), or transfected with siRNAs targeting the 3 inducible iP subunits (Arm C; n = 5). RESULTS: Vaccination stimulated antigen-specific T cell responses in all subjects, which peaked after 3-4 vaccinations, but remained elevated in Arm C subjects. Also in Arm C, circulating melanoma cell levels (as detected by quantitative PCR) fell, and T cell lytic activity against autologous melanoma was induced. In HLA-A2⁺ subjects, CD8⁺ T cells that bound tetramers loaded with cP-derived melanoma antigenic peptides were found in the peripheral blood only in Arm C subjects. Of 2 subjects with active disease (both in Arm C), one had a partial clinical response, while the other, who exhibited diffuse dermal and soft tissue metastases, had a complete response. CONCLUSION: These results suggest that the efficacy of melanoma DC-based immunotherapy is enhanced when tumor antigen-loaded DCs used for vaccination express cPs. TRIAL REGISTRATION: Clinicaltrials.gov NCT00672542. FUNDING: Duke Clinical Research Institute/Duke Translational Medicine Institute, Duke Melanoma Consortium, and Duke University Department of Surgery.


Assuntos
Células Dendríticas/transplante , Melanoma/terapia , Complexo de Endopeptidases do Proteassoma/metabolismo , Idoso , Idoso de 80 Anos ou mais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer , Células Dendríticas/enzimologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imunoterapia , Metástase Linfática , Masculino , Melanoma/imunologia , Melanoma/secundário , Pessoa de Meia-Idade , Complexo de Endopeptidases do Proteassoma/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Interferente Pequeno/genética , Resultado do Tratamento , Células Tumorais Cultivadas
16.
Methods Mol Biol ; 969: 111-25, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296931

RESUMO

Transfecting with in vitro transcribed, protein-encoding mRNA is a simple yet effective method to express high levels of the desired RNA-encoded proteins in primary cells. Cells can be transfected with antigen-encoding mRNA, which is translated into protein and is processed by the cellular antigen-processing pathway to generate antigen-presenting cells. Another elegant and increasingly popular application is to transfect cells with mRNA that encodes immune modulating molecules (cytokines, chemokines, toll-like receptors (TLRs), immune receptor ligands, immune receptor targeting antibodies) which, when translated into protein, can program cell behavior and/or function. In this chapter we describe an efficient method to deliver mRNA into human dendritic cells (DCs) by electroporation. This is currently the method of choice to deliver mRNA into antigen-presenting cells for generating vaccines for cancer immunotherapy.


Assuntos
Engenharia Celular/métodos , Células Dendríticas , Expressão Gênica , RNA Mensageiro/química , Transfecção/métodos , Antígenos/biossíntese , Antígenos/genética , Antígenos/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/metabolismo , Linhagem Celular , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , RNA Mensageiro/genética , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , Receptores Toll-Like/biossíntese , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
17.
Methods Mol Biol ; 969: 101-10, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296930

RESUMO

Ex vivo activated B cells are an alternative source of antigen presenting cells (APC). However, the ability of ex vivo activated B cells to function as potent APCs has been a concern especially when compared to dendritic cells (DC). Herein, we introduce a strategy to modulate antigen presentation and immune stimulation functions of activated B cells by co-transfection with multiple mRNAs encoding costimulatory molecules (OX40L, 4-1BBL, and CD80), cytokines (IL-12p35 and IL-12p40) and antigen. These activated B cells modified to express immune stimulatory molecules can be a potent alternative to DCs in immunotherapy.


Assuntos
Células Apresentadoras de Antígenos , Linfócitos B , Engenharia Celular/métodos , RNA Mensageiro/química , Transfecção/métodos , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos/genética , Antígenos/imunologia , Antígenos/metabolismo , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos CD/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Humanos , Subunidade p35 da Interleucina-12/biossíntese , Subunidade p35 da Interleucina-12/genética , Subunidade p35 da Interleucina-12/imunologia , Subunidade p40 da Interleucina-12/biossíntese , Subunidade p40 da Interleucina-12/genética , Subunidade p40 da Interleucina-12/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia
18.
PLoS One ; 8(1): e53150, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23341929

RESUMO

The forkhead transcription factor FOXP3 is necessary for induction of regulatory T lymphocytes (Tregs) and their immunosuppressive function. We have previously demonstrated that targeting Tregs by vaccination of mice with murine FOXP3 mRNA-transfected dendritic cells (DCs) elicits FOXP3-specific T cell responses and enhances tumor immunity. It is clear that FOXP3 expression is not restricted to T-cell lineage and herein, using RT-PCR, flow cytometry, and western immunoblot we demonstrate for the first time that FOXP3 is expressed in inflammatory breast cancer (IBC) cells, SUM149 (triple negative, ErbB1-activated) and SUM190 (ErbB2-overexpressing). Importantly, FOXP3-specific T cells generated in vitro using human FOXP3 RNA-transfected DCs as stimulators efficiently lyse SUM149 cells. Interestingly, an isogenic model (rSUM149) derived from SUM149 with an enhanced anti-apoptotic phenotype was resistant to FOXP3-specific T cell mediated lysis. The MHC class I cellular processing mechanism was intact in both cell lines at the protein and transcription levels suggesting that the resistance to cytolysis by rSUM149 cells was not related to MHC class I expression or to the MHC class I antigen processing machinery in these cells. Our data suggest that FOXP3 may be an effective tumor target in IBC cells however increased anti-apoptotic signaling can lead to immune evasion.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Neoplasias Inflamatórias Mamárias/imunologia , Neoplasias Inflamatórias Mamárias/patologia , Animais , Apresentação de Antígeno/imunologia , Apoptose/imunologia , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citotoxicidade Imunológica , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Ativação Linfocitária/imunologia , Camundongos , Fenótipo , Recidiva , Proteínas Quinases Associadas a Fase S/metabolismo , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
19.
Eur J Immunol ; 41(12): 3553-63, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22028176

RESUMO

Cancer vaccines have now demonstrated clinical efficacy, but immune modulatory mechanisms that prevent autoimmunity limit their effectiveness. Systemic administration of mAbs targeting the immune modulatory receptors CTLA-4 and glucocorticoid-induced TNFR-related protein (GITR) on Treg and effector T cells augments anti-tumor immunity both experimentally and clinically, but can induce life-threatening autoimmunity. We hypothesized that local delivery of anti-CTLA-4 and anti-GITR mAbs to the sites where T cells and tumor antigen-loaded DC vaccines interact would enhance the induction of anti-tumor immunity while avoiding autoimmunity. To achieve this goal, DCs transfected with mRNA encoding the H and L chains of anti-mouse CTLA-4 and GITR mAbs were co-administered with tumor antigen mRNA-transfected DCs. We observed enhanced induction of anti-tumor immunity and significantly improved survival in melanoma-bearing mice, without signs of autoimmunity. Using in vitro assays with human DCs, we demonstrated that DCs transfected with mRNA encoding a humanized anti-CTLA-4 mAb and mRNA encoding a soluble human GITR-L fusion protein enhance the induction of anti-tumor CTLs in response to DCs transfected with mRNAs encoding either melanoma or breast cancer antigens. Based on these results, this approach of using local delivery of immune modulators to enhance vaccine-induced immunity is currently being evaluated in a phase I clinical cancer immunotherapy trial.


Assuntos
Antígeno CTLA-4/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Melanoma Experimental/imunologia , Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Autoimunidade/imunologia , Células CHO , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Vacinas Anticâncer/genética , Vacinas Anticâncer/metabolismo , Linhagem Celular Tumoral , Cricetinae , Células Dendríticas/metabolismo , Feminino , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/metabolismo , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/imunologia , Cadeias Leves de Imunoglobulina/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transfecção/métodos , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/metabolismo
20.
Expert Opin Biol Ther ; 10(4): 563-74, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20128707

RESUMO

IMPORTANCE OF THE FIELD: Although studies have demonstrated that antigen-loaded dendritic cells (DC) elicit antigen-specific immune responses, the clinical benefit from DC-based cancer immunotherapy remains low. RNA, in the form of mRNA, has not only been used as a source of antigen but more recently as a way to stimulate DC to produce immunostimulatory molecules. As siRNA it has allowed researchers to modify DC to produce a favorable cytokine profile or to present antigen that may generate the desired immune response. AREAS COVERED IN THIS REVIEW: When loading DC with RNA that encodes immunostimulatory protein, rather than a source of antigen, optimal translation and efficient transfection into DC are critical. Studies addressing these issues and the functional consequences of modulating DC function are reviewed. WHAT THE READER WILL GAIN: RNA can be used to load DC with antigen and to encode proteins that will enhance the immune response. Co-transfection with multiple mRNAs or mRNA plus siRNA can significantly improve vaccine efficacy. TAKE HOME MESSAGE: One conclusion from Phase I clinical trials with DC loaded with tumor antigen is that tumor-specific induction of immune responses is not sufficient to destroy pre-established tumors. The advantage of transfection with mRNA is the ability to load DC with antigen-encoding mRNA and immunostimulatory protein-encoding mRNA to achieve the desired clinical response.


Assuntos
Células Dendríticas/imunologia , RNA/administração & dosagem , Antígenos/imunologia , Movimento Celular , Ensaios Clínicos Fase I como Assunto , Humanos , Biossíntese de Proteínas , RNA/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...