Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cancer Immunol Immunother ; 66(12): 1589-1595, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28825123

RESUMO

BACKGROUND: We evaluated circulating levels of immunosuppressive regulatory T cells (Tregs) and other lymphocyte subsets in patients with newly diagnosed medulloblastoma (MBL) undergoing surgery compared to a control cohort of patients undergo craniectomy for correction of Chiari malformation (CM) and further determined the impact of standard irradiation and chemotherapy on this cell population. METHODS: Eligibility criteria for this biologic study included age 4-21 years, patients with CM undergoing craniectomy (as non-malignant surgical controls) and receiving dexamethasone for prevention of post-operative nausea, and those with newly diagnosed posterior fossa tumors (PFT) undergoing surgical resection and receiving dexamethasone as an anti-edema measure. Patients with confirmed MBL were also followed for longitudinal blood collection and analysis during radiotherapy and chemotherapy. RESULTS: A total of 54 subjects were enrolled on the study [22-CM, 18-MBL, and 14-PFT]. Absolute number and percentage Tregs (defined as CD4+CD25+FoxP3+CD127low/-) at baseline were decreased in MBL and PFT compared to CM [p = 0.0016 and 0.001, respectively). Patients with MBL and PFT had significantly reduced overall CD4+ T cell count (p = 0.0014 and 0.0054, respectively) compared to those with CM. Radiation and chemotherapy treatment in patients with MBL reduced overall lymphocyte counts; however, within the CD4+ T cell compartment, Tregs increased during treatment but gradually declined post therapy. CONCLUSIONS: Our results demonstrate that patients with MBL and PFT exhibit overall reduced CD4+ T cell counts at diagnosis but not an elevated proportion of Tregs. Standard treatment exacerbates lymphopenia in those with MBL while enriching for immunosuppressive Tregs over time.


Assuntos
Neoplasias Cerebelares/imunologia , Neoplasias Cerebelares/terapia , Meduloblastoma/imunologia , Meduloblastoma/terapia , Linfócitos T Reguladores/imunologia , Adolescente , Adulto , Neoplasias Cerebelares/sangue , Quimiorradioterapia , Criança , Pré-Escolar , Craniotomia , Feminino , Humanos , Masculino , Meduloblastoma/sangue , Adulto Jovem
2.
Cancer Immunol Immunother ; 65(2): 205-11, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26759007

RESUMO

Regulatory B cells that secrete IL-10 (IL-10(+) Bregs) represent a suppressive subset of the B cell compartment with prominent anti-inflammatory capacity, capable of suppressing cellular and humoral responses to cancer and vaccines. B lymphocyte stimulator (BLyS) is a key regulatory molecule in IL-10(+) Breg biology with tightly controlled serum levels. However, BLyS levels can be drastically altered upon chemotherapeutic intervention. We have previously shown that serum BLyS levels are elevated, and directly associated, with increased antigen-specific antibody titers in patients with glioblastoma (GBM) undergoing lymphodepletive temozolomide chemotherapy and vaccination. In this study, we examined corresponding IL-10(+) Breg responses within this patient population and demonstrate that the IL-10(+) Breg compartment remains constant before and after administration of the vaccine, despite elevated BLyS levels in circulation. IL-10(+) Breg frequencies were not associated with serum BLyS levels, and ex vivo stimulation with a physiologically relevant concentration of BLyS did not increase IL-10(+) Breg frequency. However, BLyS stimulation did increase the frequency of the overall B cell compartment and promoted B cell proliferation upon B cell receptor engagement. Therefore, using BLyS as an adjuvant with therapeutic peptide vaccination could promote humoral immunity with no increase in immunosuppressive IL-10(+) Bregs. These results have implications for modulating humoral responses in human peptide vaccine trials in patients with GBM.


Assuntos
Fator Ativador de Células B/sangue , Linfócitos B Reguladores/imunologia , Glioblastoma/sangue , Glioblastoma/imunologia , Contagem de Linfócitos , Anticorpos/sangue , Anticorpos/imunologia , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Linfócitos B Reguladores/metabolismo , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Glioblastoma/mortalidade , Glioblastoma/terapia , Humanos , Imunoterapia/métodos , Interleucina-10/metabolismo , Ativação Linfocitária/imunologia , Temozolomida , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
3.
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
4.
Cancer Immunol Immunother ; 62(6): 983-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23591978

RESUMO

B lymphocyte stimulator (BLyS) is a cytokine involved in differentiation and survival of follicular B cells along with humoral response potentiation. Lymphopenia is known to precipitate dramatic elevation in serum BLyS; however, the use of this effect to enhance humoral responses following vaccination has not been evaluated. We evaluated BLyS serum levels and antigen-specific antibody titers in 8 patients undergoing therapeutic temozolomide (TMZ)-induced lymphopenia, with concomitant vaccine against a tumor-specific mutation in the epidermal growth factor receptor (EGFRvIII). Our studies demonstrate that TMZ-induced lymphopenia corresponded with spikes in serum BLyS that directly preceded the induction of anti-EGFRvIII antigen-specific antibody titers, in some cases as high as 1:2,000,000. Our data are the first clinical observation of BLyS serum elevation and greatly enhanced humoral immune responses as a consequence of chemotherapy-induced lymphopenia. These observations should be considered for the development of future vaccination strategies in the setting of malignancy.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Fator Ativador de Células B/sangue , Vacinas Anticâncer/imunologia , Dacarbazina/análogos & derivados , Glioblastoma/imunologia , Depleção Linfocítica , Anticorpos/sangue , Especificidade de Anticorpos/imunologia , Antineoplásicos Alquilantes/efeitos adversos , Dacarbazina/efeitos adversos , Dacarbazina/uso terapêutico , Glioblastoma/terapia , Humanos , Linfopenia/sangue , Linfopenia/induzido quimicamente , Temozolomida
5.
Blood ; 118(11): 3003-12, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21768296

RESUMO

Lymphodepletion augments adoptive cell transfer during antitumor immunotherapy, producing dramatic clinical responses in patients with malignant melanoma. We report that the lymphopenia induced by the chemotherapeutic agent temozolomide (TMZ) enhances vaccine-driven immune responses and significantly reduces malignant growth in an established model of murine tumorigenesis. Unexpectedly, despite the improved antitumor efficacy engendered by TMZ-induced lymphopenia, there was a treatment related increase in the frequency of immunosuppressive regulatory T cells (T(Regs); P = .0006). Monoclonal antibody (mAb)-mediated inhibition of the high-affinity IL-2 receptor α (IL-2Rα/CD25) during immunotherapy in normal mice depleted T(Regs) (73% reduction; P = .0154) but also abolished vaccine-induced immune responses. However, during lymphodepletion, IL-2Rα blockade decreased T(Regs) (93% reduction; P = .0001) without impairing effector T-cell responses, to augment therapeutic antitumor efficacy (66% reduction in tumor growth; P = .0024). Of clinical relevance, we also demonstrate that anti-IL-2Rα mAb administration during recovery from lymphodepletive TMZ in patients with glioblastoma reduced T(Reg) frequency (48% reduction; P = .0061) while permitting vaccine-stimulated antitumor effector cell expansion. To our knowledge, this is the first report of systemic antibody-mediated T(Reg) depletion during lymphopenia and the consequent synergistic enhancement of vaccine-driven cellular responses, as well as the first demonstration that anti-IL-2Rα mAbs function differentially in nonlymphopenic versus lymphopenic contexts.


Assuntos
Anticorpos Monoclonais/farmacologia , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Depleção Linfocítica/métodos , Linfopenia/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Adulto , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Vacinas Anticâncer/uso terapêutico , Células Cultivadas , Terapia Combinada , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Daclizumabe , Avaliação Pré-Clínica de Medicamentos , Glioblastoma/imunologia , Glioblastoma/terapia , Humanos , Imunoglobulina G/farmacologia , Imunoglobulina G/uso terapêutico , Imunoterapia/métodos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Linfopenia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Temozolomida , Adulto Jovem
6.
Clin Cancer Res ; 14(18): 5759-68, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18794085

RESUMO

PURPOSE: Activation of signal transducers and activators of transcription 3 (STAT3) has been identified as a central mediator of melanoma growth and metastasis. We hypothesized that WP1066, a novel STAT3 blockade agent, has marked antitumor activity, even against the melanoma metastasis to brain, a site typically refractory to therapies. EXPERIMENTAL DESIGN: The antitumor activities and related mechanisms of WP1066 were investigated both in vitro on melanoma cell lines and in vivo on mice with subcutaneously syngeneic melanoma or with intracerebral melanoma tumors. RESULTS: WP1066 achieved an IC(50) of 1.6, 2.3, and 1.5 mumol/L against melanoma cell line A375, B16, and B16EGFRvIII, respectively. WP1066 suppressed the phosphorylation of Janus-activated kinase 2 and STAT3 (Tyr705) in these cells. Tumor growth in mice with subcutaneously established syngeneic melanoma was markedly inhibited by WP1066 compared with that in controls. Long-term survival (>78 days) was observed in 80% of mice with established intracerebral syngeneic melanoma treated with 40 mg/kg of WP1066 in contrast to control mice who survived for a median of 15 days. Although WP1066 did not induce immunologic memory or enhance humoral responses to EGFRvIII, this compound reduced the production of immunosuppressive cytokines and chemokines (transforming growth factor-beta, RANTES, MCP-1, vascular endothelial growth factor), markedly inhibited natural and inducible Treg proliferation, and significantly increased cytotoxic immune responses of T cells. CONCLUSIONS: The antitumor cytotoxic effects of WP1066 and its ability to induce antitumor immune responses suggest that this compound has potential for the effective treatment of melanoma metastatic to brain.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Piridinas/uso terapêutico , Fator de Transcrição STAT3/antagonistas & inibidores , Linfócitos T Reguladores/efeitos dos fármacos , Tirfostinas/uso terapêutico , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Piridinas/farmacologia , Tirfostinas/farmacologia
7.
Neuro Oncol ; 10(1): 10-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17951512

RESUMO

Human cytomegalovirus (HCMV) has been described to be associated with several human malignancies, though the frequency of detection remains controversial. It is unclear whether HCMV plays an active role in malignant tumor progression or becomes reactivated under pathologic conditions that result in chronic inflammation or immunosuppression. In this study, we report on the investigation of detecting HCMV in the tumors and peripheral blood of patients with newly diagnosed glioblastoma multiforme (GBM). Using immunohistochemistry, in situ hybridization, and polymerase chain reaction amplification of viral DNA, the detection of HCMV was investigated in tumor and blood specimens from patients with GBM as well as in the peripheral blood of normal volunteers and patients undergoing craniotomy for diagnoses other than GBM. We found that a high percentage (>90%) of GBM tumors, not surrounding normal brain, are associated with HCMV nucleic acids and proteins. Furthermore, a significant proportion of patients (80%) with newly diagnosed GBM have detectable HCMV DNA in their peripheral blood, while sero-positive normal donors and other surgical patients did not exhibit detectable virus, suggesting either a systemic reactivation of HCMV within patients with GBM or shedding of viral DNA from infected tumor cells into the periphery. These results confirm the association of HCMV with malignant gliomas and demonstrate that subclinical HCMV viremia (presence of viral DNA in blood without clinical symptoms of infection) is a previously unrecognized disease spectrum in patients with GBM.


Assuntos
Neoplasias Encefálicas/virologia , Infecções por Citomegalovirus/complicações , Glioblastoma/virologia , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/epidemiologia , DNA Viral/análise , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Viremia/complicações , Viremia/epidemiologia
8.
J Immunol Methods ; 339(1): 74-81, 2008 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18775433

RESUMO

The epidermal growth factor receptor variant III (EGFRvIII) is a consistent tumor-specific mutation that is widely expressed in glioblastoma multiforme (GBM) and other neoplasms. As such it represents a truly tumor-specific target for antitumor immunotherapy. Although endogenous humoral responses to EGFRvIII have been reported in patients with EGFRvIII-expressing breast cancer, it is not known whether de novo responses can be generated or endogenous responses enhanced with an EGFRvIII-specific vaccine. To assess this in clinical trials, we have developed and validated an immunoassay to measure and isolate anti-EGFRvIII and anti-KLH antibodies from the serum of patients vaccinated with an EGFRvIII-specific peptide (PEPvIII) conjugated to keyhole limpet hemocyanin (KLH). Using magnetic beads with immobilized antigen we captured and detected anti-EGFRvIII and anti-KLH antibodies in serum from patients before and after vaccinations. Using this assay, we found that significant levels of antibody for tumor-specific antigen EGFRvIII (>4 microg/mL) and KLH could be induced after vaccination with PEPvIII-KLH.


Assuntos
Anticorpos Antineoplásicos/sangue , Formação de Anticorpos , Vacinas Anticâncer/uso terapêutico , Glioblastoma/sangue , Hemocianinas/uso terapêutico , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/uso terapêutico , Adjuvantes Imunológicos , Anticorpos Antineoplásicos/imunologia , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Vacinas Anticâncer/imunologia , Feminino , Glioblastoma/imunologia , Glioblastoma/terapia , Hemocianinas/imunologia , Humanos , Masculino , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/imunologia , Vacinação/métodos
9.
J Immunol Methods ; 459: 90-93, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29859231

RESUMO

The ELISpot assay prevails as one of the most sensitive and meaningful assays for the detection of antigen-specific, effector immune responses. Acquisition of cellular analyte for ELISpot analysis is typically not problematic when derived from tissues enriched in lymphocytes (e.g., lymphoid organs and blood); however, cell processing becomes more difficult when lymphocytes represent only a very minor population relative to the source tissue, especially when the source tissue is in limited supply (e.g., small mouse tumors). Traditional enzymatic-based methods for dissociating tumors often result in poor yields, inconsistent lymphocyte enrichment, and can have deleterious effects on lymphocyte phenotype and function. To address these limitations, we have developed an enzyme-free protocol for processing tumor infiltrating lymphocytes (TILs) from small mouse tumors, which enables the enumeration of antigen-specific effector lymphocytes using ELISpot analysis. This procedure is predicated on the dissociation of tumor tissue using gentle agitation with a paddle blender followed by a brief in vitro culture period to remove adherent cells, as well as to revive lymphocytes from a non-responsive state. Although this method is demonstrated with mouse intracerebral tumors, we have found that this protocol is applicable to peripheral tumors and may likely extend to alternative tissue sources wherein lymphocytes exist in low numbers.


Assuntos
Separação Celular/métodos , ELISPOT , Linfócitos do Interstício Tumoral/citologia , Neoplasias/imunologia , Linfócitos T/citologia , Animais , Enzimas , Camundongos , Camundongos Endogâmicos C57BL
10.
Clin Cancer Res ; 24(15): 3611-3631, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29703821

RESUMO

Purpose: Conventional therapy for malignant glioma fails to specifically target tumor cells. In contrast, substantial evidence indicates that if appropriately redirected, T cells can precisely eradicate tumors. Here we report the rational development of a fully human bispecific antibody (hEGFRvIII-CD3 bi-scFv) that redirects human T cells to lyse malignant glioma expressing a tumor-specific mutation of the EGFR (EGFRvIII).Experimental Design: We generated a panel of bispecific single-chain variable fragments and optimized design through successive rounds of screening and refinement. We tested the ability of our lead construct to redirect naïve T cells and induce target cell-specific lysis. To test for efficacy, we evaluated tumor growth and survival in xenogeneic and syngeneic models of glioma. Tumor penetrance following intravenous drug administration was assessed in highly invasive, orthotopic glioma models.Results: A highly expressed bispecific antibody with specificity to CD3 and EGFRvIII was generated (hEGFRvIII-CD3 bi-scFv). Antibody-induced T-cell activation, secretion of proinflammatory cytokines, and proliferation was robust and occurred exclusively in the presence of target antigen. hEGFRvIII-CD3 bi-scFv was potent and target-specific, mediating significant lysis of multiple malignant glioma cell lines and patient-derived malignant glioma samples that heterogeneously express EGFRvIII. In both subcutaneous and orthotopic models, well-engrafted, patient-derived malignant glioma was effectively treated despite heterogeneity of EGFRvIII expression; intravenous hEGFRvIII-CD3 bi-scFv administration caused significant regression of tumor burden (P < 0.0001) and significantly extended survival (P < 0.0001). Similar efficacy was obtained in highly infiltrative, syngeneic glioma models, and intravenously administered hEGFRvIII-CD3 bi-scFv localized to these orthotopic tumors.Conclusions: We have developed a clinically translatable bispecific antibody that redirects human T cells to safely and effectively treat malignant glioma. On the basis of these results, we have developed a clinical study of hEGFRvIII-CD3 bi-scFv for patients with EGFRvIII-positive malignant glioma. Clin Cancer Res; 24(15); 3611-31. ©2018 AACR.


Assuntos
Complexo CD3/antagonistas & inibidores , Receptores ErbB/antagonistas & inibidores , Glioma/tratamento farmacológico , Imunoterapia , Animais , Anticorpos Biespecíficos/farmacologia , Complexo CD3/imunologia , Linhagem Celular Tumoral , Receptores ErbB/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Glioma/imunologia , Glioma/patologia , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Masculino , Camundongos , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Res ; 78(1): 256-264, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29093005

RESUMO

Median survival for glioblastoma (GBM) remains <15 months. Human cytomegalovirus (CMV) antigens have been identified in GBM but not normal brain, providing an unparalleled opportunity to subvert CMV antigens as tumor-specific immunotherapy targets. A recent trial in recurrent GBM patients demonstrated the potential clinical benefit of adoptive T-cell therapy (ATCT) of CMV phosphoprotein 65 (pp65)-specific T cells. However, ex vivo analyses from this study found no change in the capacity of CMV pp65-specific T cells to gain multiple effector functions or polyfunctionality, which has been associated with superior antitumor efficacy. Previous studies have shown that dendritic cells (DC) could further enhance tumor-specific CD8+ T-cell polyfunctionality in vivo when administered as a vaccine. Therefore, we hypothesized that vaccination with CMV pp65 RNA-loaded DCs would enhance the frequency of polyfunctional CMV pp65-specific CD8+ T cells after ATCT. Here, we report prospective results of a pilot trial in which 22 patients with newly diagnosed GBM were initially enrolled, of which 17 patients were randomized to receive CMV pp65-specific T cells with CMV-DC vaccination (CMV-ATCT-DC) or saline (CMV-ATCT-saline). Patients who received CMV-ATCT-DC vaccination experienced a significant increase in the overall frequencies of IFNγ+, TNFα+, and CCL3+ polyfunctional, CMV-specific CD8+ T cells. These increases in polyfunctional CMV-specific CD8+ T cells correlated (R = 0.7371, P = 0.0369) with overall survival, although we cannot conclude this was causally related. Our data implicate polyfunctional T-cell responses as a potential biomarker for effective antitumor immunotherapy and support a formal assessment of this combination approach in a larger randomized study.Significance: A randomized pilot trial in patients with GBM implicates polyfunctional T-cell responses as a biomarker for effective antitumor immunotherapy. Cancer Res; 78(1); 256-64. ©2017 AACR.


Assuntos
Neoplasias Encefálicas/terapia , Células Dendríticas/imunologia , Glioblastoma/terapia , Imunoterapia Adotiva/métodos , Linfócitos T/imunologia , Transferência Adotiva , Adulto , Idoso , Linfócitos T CD8-Positivos/imunologia , Citomegalovirus , Células Dendríticas/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/metabolismo , Linfócitos T/transplante , Resultado do Tratamento , Proteínas da Matriz Viral/metabolismo
12.
J Neuroimmunol ; 182(1-2): 63-72, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17137636

RESUMO

For the study of malignant glioma, we have previously characterized a highly tumorigenic murine astrocytoma, SMA-560, which arose spontaneously in an inbred, immunocompetent VM/Dk mouse. Using this cell line as a model of murine glioma, we performed DNA microarray analysis of autologous normal murine astroctyes (NMA) and SMA-560 tumor cells grown in monolayer culture or intracranially in syngeneic immunocompetent or immunocompromised hosts in order to determine whether tumors grown in vitro recreate the complex genetic regulation that occurs in vivo. Our findings support our hypothesis that glioma phenotype in vitro may be quite different in vivo and significantly altered by in situ growth factors and other invading cell populations.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Expressão Gênica , Glioma/genética , Glioma/imunologia , Imunidade/genética , Animais , Astrócitos/metabolismo , Astrocitoma/genética , Astrocitoma/imunologia , Astrocitoma/metabolismo , Astrocitoma/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Análise por Conglomerados , Perfilação da Expressão Gênica , Glioma/metabolismo , Glioma/patologia , Imunocompetência/genética , Hospedeiro Imunocomprometido/genética , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal , Linfócitos T/patologia
13.
Clin Cancer Res ; 12(24): 7306-15, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17189402

RESUMO

PURPOSE: Analyses of T-cell mRNA expression profiles in glioblastoma multiforme has not been previously reported but may help to define and characterize the immunosuppressed phenotype in patients with this type of cancer. EXPERIMENTAL DESIGN: We did microarray studies that have shown significant and fundamental differences in the expression profiles of CD4(+) and CD8(+) T cells and immunosuppressive CD4(+)CD25(+)CD45RO(+)FoxP3(+) regulatory T cells (T(reg)) from normal healthy volunteers compared with patients with newly diagnosed glioblastoma multiforme. For these investigations, we isolated total RNA from enriched CD4(+) and CD8(+) T cell or T(reg) cell populations from age-matched individuals and did microarray analyses. RESULTS: ANOVA and principal components analysis show that the various T cell compartments exhibit consistently similar mRNA expression profiles among individuals within either healthy or brain tumor groups but reflect significant differences between these groups. Compared with healthy volunteers, CD4(+) and CD8(+) T cells from patients with glioblastoma multiforme display coordinate down-regulation of genes involved in T cell receptor ligation, activation, and intracellular signaling. In contrast, T(regs) from patients with glioblastoma multiforme exhibit increased levels of transcripts involved in inhibiting host immunity. CONCLUSION: Our findings support the notion that key differences between expression profiles in T-cell populations from patients with glioblastoma multiforme results from differential expression of the immunologic transcriptome, such that a limited number of genes are principally important in producing the dysregulated T-cell phenotype.


Assuntos
Neoplasias Encefálicas/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Glioma/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Linfócitos T/metabolismo , Adulto , Estudos de Casos e Controles , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Humanos , Imunofenotipagem/métodos , Masculino , Pessoa de Meia-Idade
14.
Clin Cancer Res ; 23(8): 1898-1909, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28411277

RESUMO

Purpose: Patients with glioblastoma have less than 15-month median survival despite surgical resection, high-dose radiation, and chemotherapy with temozolomide. We previously demonstrated that targeting cytomegalovirus pp65 using dendritic cells (DC) can extend survival and, in a separate study, that dose-intensified temozolomide (DI-TMZ) and adjuvant granulocyte macrophage colony-stimulating factor (GM-CSF) potentiate tumor-specific immune responses in patients with glioblastoma. Here, we evaluated pp65-specific cellular responses following DI-TMZ with pp65-DCs and determined the effects on long-term progression-free survival (PFS) and overall survival (OS).Experimental Design: Following standard-of-care, 11 patients with newly diagnosed glioblastoma received DI-TMZ (100 mg/m2/d × 21 days per cycle) with at least three vaccines of pp65 lysosome-associated membrane glycoprotein mRNA-pulsed DCs admixed with GM-CSF on day 23 ± 1 of each cycle. Thereafter, monthly DI-TMZ cycles and pp65-DCs were continued if patients had not progressed.Results: Following DI-TMZ cycle 1 and three doses of pp65-DCs, pp65 cellular responses significantly increased. After DI-TMZ, both the proportion and proliferation of regulatory T cells (Tregs) increased and remained elevated with serial DI-TMZ cycles. Median PFS and OS were 25.3 months [95% confidence interval (CI), 11.0-∞] and 41.1 months (95% CI, 21.6-∞), exceeding survival using recursive partitioning analysis and matched historical controls. Four patients remained progression-free at 59 to 64 months from diagnosis. No known prognostic factors [age, Karnofsky performance status (KPS), IDH-1/2 mutation, and MGMT promoter methylation] predicted more favorable outcomes for the patients in this cohort.Conclusions: Despite increased Treg proportions following DI-TMZ, patients receiving pp65-DCs showed long-term PFS and OS, confirming prior studies targeting cytomegalovirus in glioblastoma. Clin Cancer Res; 23(8); 1898-909. ©2017 AACR.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/terapia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/transplante , Glioblastoma/terapia , Fosfoproteínas/uso terapêutico , Proteínas da Matriz Viral/uso terapêutico , Adjuvantes Imunológicos , Idoso , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Terapia Combinada , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Células Dendríticas/imunologia , Intervalo Livre de Doença , Feminino , Glioblastoma/imunologia , Glioblastoma/mortalidade , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/imunologia , Linfócitos T Reguladores/imunologia , Temozolomida , Proteínas da Matriz Viral/imunologia
15.
Cancer Immunol Res ; 3(4): 320-5, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25387895

RESUMO

Therapeutic vaccination of patients with cancer-targeting tumor-associated antigens is a promising strategy for the specific eradication of invasive malignancies with minimal toxicity to normal tissues. However, as increasingly potent modalities for stimulating immunologic responses are developed for clinical evaluation, the risk of inflammatory and autoimmune toxicities also may be exacerbated. In this report, we describe the induction of a severe (grade 3) immunologic reaction in a patient with newly diagnosed glioblastoma (GBM) receiving autologous RNA-pulsed dendritic cell (DC) vaccines admixed with GM-CSF and administered coordinately with cycles of dose-intensified temozolomide. Shortly after the eighth administration of the admixed intradermal vaccine, the patient experienced dizziness, flushing, conjunctivitis, headache, and the outbreak of a disseminated macular/papular rash and bilateral indurated injection sites. Immunologic workup of patient reactivity revealed sensitization to the GM-CSF component of the vaccine and the production of high levels of anti-GM-CSF autoantibodies during vaccination. Removal of GM-CSF from the DC vaccine allowed continued vaccination without incident. Despite the known lymphodepletive and immunosuppressive effects of temozolomide, these observations demonstrate the capacity for the generation of severe immunologic reactivity in patients with GBM receiving DC-based therapy during adjuvant dose-intensified temozolomide.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Vacinas Anticâncer/efeitos adversos , Dacarbazina/análogos & derivados , Células Dendríticas/transplante , Glioblastoma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Autoanticorpos/biossíntese , Vacinas Anticâncer/uso terapêutico , Terapia Combinada , Dacarbazina/efeitos adversos , Células Dendríticas/imunologia , Glioblastoma/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Temozolomida
17.
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
18.
Clin Cancer Res ; 20(4): 972-84, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24352643

RESUMO

PURPOSE: Chimeric antigen receptor (CAR) transduced T cells represent a promising immune therapy that has been shown to successfully treat cancers in mice and humans. However, CARs targeting antigens expressed in both tumors and normal tissues have led to significant toxicity. Preclinical studies have been limited by the use of xenograft models that do not adequately recapitulate the immune system of a clinically relevant host. A constitutively activated mutant of the naturally occurring epidermal growth factor receptor (EGFRvIII) is antigenically identical in both human and mouse glioma, but is also completely absent from any normal tissues. EXPERIMENTAL DESIGN: We developed a third-generation, EGFRvIII-specific murine CAR (mCAR), and performed tests to determine its efficacy in a fully immunocompetent mouse model of malignant glioma. RESULTS: At elevated doses, infusion with EGFRvIII mCAR T cells led to cures in all mice with brain tumors. In addition, antitumor efficacy was found to be dependent on lymphodepletive host conditioning. Selective blockade with EGFRvIII soluble peptide significantly abrogated the activity of EGFRvIII mCAR T cells in vitro and in vivo, and may offer a novel strategy to enhance the safety profile for CAR-based therapy. Finally, mCAR-treated, cured mice were resistant to rechallenge with EGFRvIII(NEG) tumors, suggesting generation of host immunity against additional tumor antigens. CONCLUSION: All together, these data support that third-generation, EGFRvIII-specific mCARs are effective against gliomas in the brain and highlight the importance of syngeneic, immunocompetent models in the preclinical evaluation of tumor immunotherapies.


Assuntos
Transferência Adotiva , Astrocitoma/terapia , Neoplasias Encefálicas/terapia , Receptores ErbB/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Animais , Astrocitoma/imunologia , Astrocitoma/metabolismo , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/imunologia , Humanos , Camundongos , Transplante de Neoplasias , Anticorpos de Cadeia Única/metabolismo , Linfócitos T/imunologia , Linfócitos T/transplante
19.
PLoS One ; 8(3): e59082, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23527092

RESUMO

Temozolomide (TMZ) is an alkylating agent shown to prolong survival in patients with high grade glioma and is routinely used to treat melanoma brain metastases. A prominent side effect of TMZ is induction of profound lymphopenia, which some suggest may be incompatible with immunotherapy. Conversely, it has been proposed that recovery from chemotherapy-induced lymphopenia may actually be exploited to potentiate T-cell responses. Here, we report the first demonstration of TMZ as an immune host-conditioning regimen in an experimental model of brain tumor and examine its impact on antitumor efficacy of a well-characterized peptide vaccine. Our results show that high-dose, myeloablative (MA) TMZ resulted in markedly reduced CD4(+), CD8(+) T-cell and CD4(+)Foxp3(+) TReg counts. Adoptive transfer of naïve CD8(+) T cells and vaccination in this setting led to an approximately 70-fold expansion of antigen-specific CD8(+) T cells over controls. Ex vivo analysis of effector functions revealed significantly enhanced levels of pro-inflammatory cytokine secretion from mice receiving MA TMZ when compared to those treated with a lower lymphodepletive, non-myeloablative (NMA) dose. Importantly, MA TMZ, but not NMA TMZ was uniquely associated with an elevation of endogenous IL-2 serum levels, which we also show was required for optimal T-cell expansion. Accordingly, in a murine model of established intracerebral tumor, vaccination-induced immunity in the setting of MA TMZ-but not lymphodepletive, NMA TMZ-led to significantly prolonged survival. Overall, these results may be used to leverage the side-effects of a clinically-approved chemotherapy and should be considered in future study design of immune-based treatments for brain tumors.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Dacarbazina/análogos & derivados , Animais , Antígenos/imunologia , Antineoplásicos Alquilantes/efeitos adversos , Neoplasias Encefálicas/mortalidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Dacarbazina/efeitos adversos , Dacarbazina/farmacologia , Modelos Animais de Doenças , Imunoterapia , Interleucina-2/sangue , Interleucina-2/farmacologia , Depleção Linfocítica , Linfopenia/induzido quimicamente , Camundongos , Camundongos Transgênicos , Temozolomida , Vacinas de Subunidades Antigênicas/imunologia
20.
PLoS One ; 7(2): e31046, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22383993

RESUMO

BACKGROUND: Preclinical studies in mice have demonstrated that the prophylactic depletion of immunosuppressive regulatory T-cells (T(Regs)) through targeting the high affinity interleukin-2 (IL-2) receptor (IL-2Rα/CD25) can enhance anti-tumor immunotherapy. However, therapeutic approaches are complicated by the inadvertent inhibition of IL-2Rα expressing anti-tumor effector T-cells. OBJECTIVE: To determine if changes in the cytokine milieu during lymphopenia may engender differential signaling requirements that would enable unarmed anti-IL-2Rα monoclonal antibody (MAbs) to selectively deplete T(Regs) while permitting vaccine-stimulated immune responses. METHODOLOGY: A randomized placebo-controlled pilot study was undertaken to examine the ability of the anti-IL-2Rα MAb daclizumab, given at the time of epidermal growth factor receptor variant III (EGFRvIII) targeted peptide vaccination, to safely and selectively deplete T(Regs) in patients with glioblastoma (GBM) treated with lymphodepleting temozolomide (TMZ). RESULTS AND CONCLUSIONS: Daclizumab treatment (n = 3) was well-tolerated with no symptoms of autoimmune toxicity and resulted in a significant reduction in the frequency of circulating CD4+Foxp3+ TRegs in comparison to saline controls (n = 3)( p = 0.0464). A significant (p<0.0001) inverse correlation between the frequency of TRegs and the level of EGFRvIII specific humoral responses suggests the depletion of TRegs may be linked to increased vaccine-stimulated humoral immunity. These data suggest this approach deserves further study. TRIAL REGISTRATION: ClinicalTrials.gov NCT00626015.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Linfopenia/metabolismo , Linfócitos T Reguladores/metabolismo , Adulto , Idoso , Anticorpos Monoclonais Humanizados/farmacologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Daclizumabe , Feminino , Humanos , Sistema Imunitário , Imunoglobulina G/farmacologia , Imunossupressores/farmacologia , Subunidade alfa de Receptor de Interleucina-2/química , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Linfócitos T/citologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa