Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 27(24): 6696-6708, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34667029

RESUMO

PURPOSE: This proof-of-principle clinical trial evaluated whether an allogeneic multiple myeloma GM-CSF-secreting vaccine (MM-GVAX) in combination with lenalidomide could deepen the clinical response in patients with multiple myeloma in sustained near complete remission (nCR). PATIENTS AND METHODS: Fifteen patients on lenalidomide were treated with MM-GVAX and pneumococcal conjugate vaccine (PCV; Prevnar) at 1, 2, 3, and 6 months. RESULTS: Eight patients (53.3%) achieved a true CR. With a median follow-up of 5 years, the median progression-free survival had not been reached, and the median overall survival was 7.8 years from enrollment. MM-GVAX induced clonal T-cell expansion and measurable cytokine responses that persisted up to 7 years in all patients. At baseline, a higher minimal residual disease was predictive of early relapse. After vaccination, a lack of both CD27-DNAM1-CD8+ T cells and antigen-presenting cells was associated with disease progression. CONCLUSIONS: MM-GVAX, along with lenalidomide, effectively primed durable immunity and resulted in long-term disease control, as suggested by the reappearance of a detectable, fluctuating M-spike without meeting the criteria for clinical relapse. For patients in a nCR, MM-GVAX administration was safe and resulted in prolonged clinical responses.


Assuntos
Vacinas Anticâncer , Transplante de Células-Tronco Hematopoéticas , Mieloma Múltiplo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfócitos T CD8-Positivos , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Humanos , Lenalidomida , Mieloma Múltiplo/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico
3.
Nat Commun ; 9(1): 741, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29467463

RESUMO

A majority of cancers fail to respond to immunotherapy with antibodies targeting immune checkpoints, such as cytotoxic T-lymphocyte antigen-4 (CTLA-4) or programmed death-1 (PD-1)/PD-1 ligand (PD-L1). Cancers frequently express transforming growth factor-ß (TGFß), which drives immune dysfunction in the tumor microenvironment by inducing regulatory T cells (Tregs) and inhibiting CD8+ and TH1 cells. To address this therapeutic challenge, we invent bifunctional antibody-ligand traps (Y-traps) comprising an antibody targeting CTLA-4 or PD-L1 fused to a TGFß receptor II ectodomain sequence that simultaneously disables autocrine/paracrine TGFß in the target cell microenvironment (a-CTLA4-TGFßRIIecd and a-PDL1-TGFßRIIecd). a-CTLA4-TGFßRIIecd is more effective in reducing tumor-infiltrating Tregs and inhibiting tumor progression compared with CTLA-4 antibody (Ipilimumab). Likewise, a-PDL1-TGFßRIIecd exhibits superior antitumor efficacy compared with PD-L1 antibodies (Atezolizumab or Avelumab). Our data demonstrate that Y-traps counteract TGFß-mediated differentiation of Tregs and immune tolerance, thereby providing a potentially more effective immunotherapeutic strategy against cancers that are resistant to current immune checkpoint inhibitors.


Assuntos
Anticorpos/uso terapêutico , Neoplasias da Mama/terapia , Imunoterapia , Melanoma/terapia , Fator de Crescimento Transformador beta/imunologia , Animais , Anticorpos/genética , Anticorpos/imunologia , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Estudos de Coortes , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Humanos , Linfócitos do Interstício Tumoral , Melanoma/genética , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos NOD , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Fator de Crescimento Transformador beta/genética
4.
PLoS One ; 12(11): e0188916, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29190813

RESUMO

Flow cytometry is the primary immunological technique used to analyze multiple parameters on complex cell populations. We present a staining method that identifies major human mononuclear lymphoid and myeloid populations (CD4+ and CD8+ T cells, γδ T cells, B cells, NK cells and monocytes), using only two fluorochromes and a minimal number of cells. Our approach increases the number of markers recordable on most flow cytometers allowing for a deeper and more comprehensive immunophenotyping.


Assuntos
Biomarcadores/metabolismo , Citometria de Fluxo/métodos , Corantes Fluorescentes/química , Linfócitos/classificação , Linhagem da Célula , Humanos , Imunofenotipagem , Linfócitos/imunologia , Mieloma Múltiplo/imunologia
5.
Front Immunol ; 7: 112, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27066005

RESUMO

The past several years have witnessed the acceptance of immunotherapy into the mainstream of therapies for patients with cancer. This has been driven by the clinical successes of antibodies to the checkpoint inhibitors, CTLA-4 and PD-1, capable of imparting long-term remissions in several solid tumors as well as Hodgkin's lymphoma (1) and the therapeutic successes of adoptive T-cell transfer with chimeric antigen receptors (2) or modified T-cell receptors (3) that have mostly utilized peripheral T-cells. One emerging area of therapeutic T cell intervention has been the utilization of marrow-infiltrating lymphocytes (MILs) - a novel form of adoptive T-cell therapy. This approach was initially developed to increase the likelihood of a precursor T-cell population with an enhanced tumor specificity in bone marrow (BM)-derived malignancies. However, the unique attributes of BM T-cells and their interaction with their microenvironment provide significant rationale to utilize these cells therapeutically in diseases that extend beyond hematologic malignancies.

6.
Cancer J ; 21(6): 501-5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26588683

RESUMO

The clinical results achieved with immunotherapy in the past few years have now firmly established it within the cancer armamentarium. Our group has explored a novel approach to adoptive T-cell therapy utilizing marrow-infiltrating lymphocytes (MILs) initially developed with the concept of utilizing a population of T cells with a higher endogenous tumor specificity. Marrow-infiltrating lymphocytes are antigen-experienced T cells that home to and remain in the bone marrow (BM) because of the unique biology of the BM microenvironment. Marrow-infiltrating lymphocytes can easily be obtained from the BM and can be expanded to demonstrate enhanced antigen specificity. Current clinical trials utilize MILs for patients with myeloma as well as patients with relapsed disease following an allogeneic transplant. Ongoing preclinical work is currently evaluating MILs for use in solid cancers as well as pediatric cancers. The examination of a MIL as a source cell for chimeric antigen receptor T or transgenic cell receptor is also in the preclinical stages. Until now, for both chimeric antigen receptor T-cell therapy and transgenic cell receptor T-cell therapy, the target cell of choice has included peripheral blood. The unique antigen-experienced properties of MILs may make them the ideal source of cell for gene modification strategies. Therefore, MILs are a distinctive set of T cells that have been shaped by the unique BM microenvironment and may play a future role as a novel immunotherapy for hematologic malignancies.


Assuntos
Medula Óssea/patologia , Imunoterapia Adotiva/métodos , Linfócitos do Interstício Tumoral/imunologia , Humanos
7.
Sci Transl Med ; 7(288): 288ra78, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25995224

RESUMO

Successful adoptive T cell therapy (ACT) requires the ability to activate tumor-specific T cells with the ability to traffic to the tumor site and effectively kill their target as well as persist over time. We hypothesized that ACT using marrow-infiltrating lymphocytes (MILs) in multiple myeloma (MM) could impart greater antitumor immunity in that they were obtained from the tumor microenvironment. We describe the results from the first clinical trial using MILs in MM. Twenty-five patients with either newly diagnosed or relapsed disease had their MILs harvested, activated and expanded, and subsequently infused on the third day after myeloablative therapy. Cells were obtained and adequately expanded in all patients with anti-CD3/CD28 beads plus interleukin-2, and a median of 9.5 × 10(8) MILs were infused. Factors indicative of response to MIL ACT included (i) the presence of measurable myeloma-specific activity of the ex vivo expanded product, (ii) low endogenous bone marrow T cell interferon-γ production at baseline, (iii) a CD8(+) central memory phenotype at baseline, and (iv) the generation and persistence of myeloma-specific immunity in the bone marrow at 1 year after ACT. Achieving at least a 90% reduction in disease burden significantly increased the progression-free survival (25.1 months versus 11.8 months; P = 0.01). This study demonstrates the feasibility and efficacy of MILs as a form of ACT with applicability across many hematologic malignancies and possibly solid tumors infiltrating the bone marrow.


Assuntos
Medula Óssea/imunologia , Imunoterapia Adotiva/métodos , Ativação Linfocitária , Linfócitos do Interstício Tumoral/transplante , Mieloma Múltiplo/terapia , Linfócitos T/transplante , Adulto , Idoso , Baltimore , Separação Celular/métodos , Células Cultivadas , Intervalo Livre de Doença , Estudos de Viabilidade , Feminino , Humanos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/mortalidade , Estimativa de Kaplan-Meier , Linfócitos do Interstício Tumoral/imunologia , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/mortalidade , Agonistas Mieloablativos/uso terapêutico , Indução de Remissão , Linfócitos T/imunologia , Fatores de Tempo , Resultado do Tratamento , Microambiente Tumoral
8.
Clin Cancer Res ; 21(1): 30-8, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25564570

RESUMO

PURPOSE: To determine if phosphodiesterase 5 (PDE5) inhibitors can augment immune function in patients with head and neck cancer through inhibition of myeloid-derived suppressor cells (MDSC). EXPERIMENTAL DESIGN: We performed a randomized, prospective, double blinded, placebo controlled, phase II clinical trial to determine the in vivo effects of systemic PDE5 inhibition on immune function in patients with head and neck squamous cell carcinoma (HNSCC). RESULTS: Tadalafil augmented immune response, increasing ex vivo T-cell expansion to a mean 2.4-fold increase compared with 1.1-fold in control patients (P = 0.01), reducing peripheral MDSC numbers to mean 0.81-fold change compared with a 1.26-fold change in control patients (P = 0.001), and increasing general immunity as measured by delayed type hypersensitivity response (P = 0.002). Tumor-specific immunity in response to HNSCC tumor lysate was augmented in tadalafil-treated patients (P = 0.04). CONCLUSIONS: These findings demonstrate that tadalafil augments general and tumor-specific immunity in patients with HNSCC and has therapeutic potential in HNSCC. Evasion of immune surveillance and suppression of systemic and tumor-specific immunity is a significant feature of head and neck cancer development. This study demonstrates that a PDE5 inhibitor, tadalafil, can reverse tumor-specific immune suppression in patients with head and neck cancer, with potential for therapeutic application.


Assuntos
Carbolinas/administração & dosagem , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Imunidade Celular/efeitos dos fármacos , Inibidores da Fosfodiesterase 5/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/imunologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/imunologia , Feminino , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma de Células Escamosas de Cabeça e Pescoço , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Tadalafila
9.
Cancer Immunol Res ; 2(8): 725-31, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24878583

RESUMO

Myeloid-derived suppressor cells (MDSC) play a significant role in tumor-induced immune suppression. Targeting their function could improve antitumor therapies. Previously, we demonstrated that phosphodiesterase 5 (PDE5) inhibition in MDSCs augmented antitumor immunity in murine models. Here, we show how the addition of the PDE5 inhibitor, tadalafil, in a patient with end-stage relapsed/refractory multiple myeloma reduced MDSC function and generated a dramatic and durable antimyeloma immune and clinical response. Strategies targeting MDSC function with PDE5 inhibitors represent a novel approach that can augment the efficacy of tumor-directed therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carbolinas/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Inibidores da Fosfodiesterase 5/uso terapêutico , Carbolinas/farmacologia , Claritromicina/administração & dosagem , Dexametasona/administração & dosagem , Humanos , Tolerância Imunológica/efeitos dos fármacos , Fatores Imunológicos/administração & dosagem , Subunidade alfa de Receptor de Interleucina-4/imunologia , Lenalidomida , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/imunologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Inibidores da Fosfodiesterase 5/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Tadalafila , Talidomida/administração & dosagem , Talidomida/análogos & derivados
10.
Cancer Res ; 64(17): 6337-43, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15342423

RESUMO

Tumor vaccines have shown promise in early clinical trials. Among them, tumor cells genetically engineered to secrete biologically active granulocyte-macrophage colony-stimulating factor (GM-CSF) can generate a systemic antitumor immune response. Although the minimal required GM-CSF dose produced by modified tumor cells to achieve a measurable antitumor effect is well known, no data examined whether an upper therapeutic limit may exist for this vaccination strategy. Because recent data demonstrate an immunosuppressive effect of GM-CSF produced by growing tumors, we thus sought to determine whether high GM-CSF doses administered in a vaccine formulation could impair antitumor immunity. Using a vaccine strategy involving a GM-CSF-producing bystander cell line (B78H1-GM) admixed with autologous tumor, we assessed the impact of varying doses of GM-CSF while maintaining a constant antigen dose. Our results defined a threshold above which a GM-CSF-based vaccine not only lost its efficacy, but more importantly for its clinical implications resulted in substantial immunosuppression in vivo. Above this threshold, GM-CSF induced Gr1+/CD11b+ myeloid suppressor cells that substantially impaired antigen-specific T-cell responses and adversely affected antitumor immune responses in vivo. The dual effects of GM-CSF are mediated by the systemic and not local concentration of this cytokine. Myeloid suppressor cell-induced immunosuppression is mediated by nitric oxide production via inducible nitric oxide synthase (iNOS) because the specific iNOS inhibitor, l-NMMA, restored antigen-specific T-cell responsiveness in vitro. Taken together, our data demonstrated the negative impact of supra-therapeutic vaccine doses of GM-CSF and underscored the importance of identifying these critical variables in an effort to increase the therapeutic efficacy of tumor vaccines.


Assuntos
Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Células Mieloides/imunologia , Neoplasias Experimentais/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Relação Dose-Resposta Imunológica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Neoplasias Experimentais/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...