Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
N Engl J Med ; 375(18): 1749-1755, 2016 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-27806233

RESUMO

Immune checkpoint inhibitors have improved clinical outcomes associated with numerous cancers, but high-grade, immune-related adverse events can occur, particularly with combination immunotherapy. We report the cases of two patients with melanoma in whom fatal myocarditis developed after treatment with ipilimumab and nivolumab. In both patients, there was development of myositis with rhabdomyolysis, early progressive and refractory cardiac electrical instability, and myocarditis with a robust presence of T-cell and macrophage infiltrates. Selective clonal T-cell populations infiltrating the myocardium were identical to those present in tumors and skeletal muscle. Pharmacovigilance studies show that myocarditis occurred in 0.27% of patients treated with a combination of ipilimumab and nivolumab, which suggests that our patients were having a rare, potentially fatal, T-cell-driven drug reaction. (Funded by Vanderbilt-Ingram Cancer Center Ambassadors and others.).


Assuntos
Anticorpos Monoclonais/efeitos adversos , Imunoterapia/efeitos adversos , Miocardite/etiologia , Miocárdio/patologia , Idoso , Anticorpos Monoclonais/uso terapêutico , Arritmias Cardíacas/induzido quimicamente , Eletrocardiografia/efeitos dos fármacos , Evolução Fatal , Feminino , Glucocorticoides/uso terapêutico , Bloqueio Cardíaco/diagnóstico , Bloqueio Cardíaco/etiologia , Humanos , Ipilimumab , Masculino , Melanoma/complicações , Melanoma/tratamento farmacológico , Pessoa de Meia-Idade , Miocardite/tratamento farmacológico , Miocardite/patologia , Miosite/induzido quimicamente , Nivolumabe
2.
Pediatr Int ; 55(2): e4-6, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23679183

RESUMO

Obstruction and thrombosis of major systemic veins can occur due to indwelling central venous catheters. If obstruction of the innominate vein or superior vena cava occurs, lymphatic drainage can be impaired due to an increase in pressure in the thoracic duct and lymphatics. We describe a case where superior vena cava syndrome, chylopericardium and chylothorax occurred in a 16-year-old girl due to an indwelling central venous catheter. This was successfully treated with removal of the line, anticoagulation and angioplasty of the innominate vein and superior vena cava.


Assuntos
Cateteres de Demora/efeitos adversos , Cateteres Venosos Centrais/efeitos adversos , Quilotórax/etiologia , Derrame Pericárdico/etiologia , Síndrome da Veia Cava Superior/etiologia , Adolescente , Quilotórax/diagnóstico , Feminino , Humanos , Síndrome de Li-Fraumeni/terapia , Derrame Pericárdico/diagnóstico , Síndrome da Veia Cava Superior/diagnóstico , Tomografia Computadorizada por Raios X , Ultrassonografia Doppler
3.
Breast Cancer Res Treat ; 134(1): 61-70, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22173696

RESUMO

Cancer progenitor cells are critical for tumor initiation and recurrence so they are an important therapeutic target. We tested whether T cells could recognize tumor antigens expressed by breast cancer progenitor cells and acquire therapeutic activity against established metastases or delay onset of spontaneous tumors. Breast tumors were derived from HER2/neu transgenic mice and propagated in vitro under conditions that selected progenitor cells which were then used as an irradiated whole cell vaccine. A minor subset of recently sensitized T cells was isolated from vaccine-draining lymph nodes then activated in vitro to achieve numerical expansion. We show that the tumor progenitor cell vaccines reversed tolerance to a known HER2/neu epitope, otherwise inhibited by Treg cells. Additional shared tumor antigens were recognized because a Neuneg subclone also induced a Th1 type immune response against breast tumors. Adoptive transfer of in vitro activated lymph node T cells-mediated regression of established metastases from multiple independently derived breast tumor lines. Moreover, adoptive transfer of effector T cells into Neu-tolerant mice, months before the onset of spontaneous tumors, significantly postponed tumor development. Interestingly, T-cell-mediated lysis of metastases stimulated an IgG response to HER2/neu as well as other shared antigens. In summary, tumor progenitor cells contain shared antigens which can lead to a cross-protective T-cell response. Moreover, antigens acquired during immune-mediated tumor destruction are presented in a manner conducive to reversal of tolerance and Ig class switching. These complementary effector mechanisms might augment therapy by eliminating refractory breast cancer stem cells.


Assuntos
Carcinoma/terapia , Imunoterapia Adotiva , Neoplasias Mamárias Experimentais/terapia , Células-Tronco Neoplásicas/imunologia , Receptor ErbB-2/genética , Animais , Antígenos CD/metabolismo , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer , Carcinoma/genética , Carcinoma/secundário , Feminino , Linfonodos/imunologia , Ativação Linfocitária , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Receptor ErbB-2/imunologia , Esferoides Celulares/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante , Células Tumorais Cultivadas
4.
J Immunol ; 184(10): 5612-8, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20382887

RESUMO

Tumor-reactive T cells can be primed in vivo, then activated in vitro to provide numerical expansion and uniform acquisiton of effector phenotype and function. Adoptive transfer of effector T cells mediates complete regression of established tumors in animal models. Some experimental models indicate that extensive in vitro proliferation of T cells inhibits efficacy and that central memory T cells (T(CM)) provide greater activity than effector memory T cells (T(EM)). Clinical studies also demonstrate that persistence of adoptively transferred T cells is associated with therapeutic response, thus identifying that conditions to maximize effector cell numbers yet retain memory function are important. In this article, we demonstrate that adoptive transfer of in vitro activated effector CD4(+) T cells into tumor-free congenic mice mediates rejection of tumor challenge 9 mo later, at which time T cells re-express activation markers and undergo rapid proliferation at tumor sites. Analysis of the phenotype of memory cells in lymphoid tissues following adoptive transfer shows high CD44 expression with heterogeneous expression of CD62L, indicating a mixture of T(EM) and T(CM) phenotypes. Memory cells were sorted into two subsets based on CD62L expression levels and then activated in vitro. Although T(EM) cells proliferated more rapidly, T(EM) and T(CM) cells acquired effector phenotype and function. These data indicate that controlled in vitro expansion of tumor-reactive T cells for adoptive immunotherapy also provides a competent memory response.


Assuntos
Antígenos de Neoplasias/administração & dosagem , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Citocinas , Memória Imunológica , Ativação Linfocitária/imunologia , Animais , Antígenos de Neoplasias/fisiologia , Linfócitos T CD4-Positivos/patologia , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/administração & dosagem , Citocinas/fisiologia , Feminino , Fibrossarcoma/imunologia , Fibrossarcoma/patologia , Fibrossarcoma/secundário , Fibrossarcoma/terapia , Imunoterapia Adotiva/métodos , Selectina L/biossíntese , Selectina L/genética , Selectina L/fisiologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias/imunologia , Transplante de Neoplasias/patologia
5.
J Immunother Cancer ; 10(4)2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35383114

RESUMO

BACKGROUND: Retrospective studies have suggested a potential risk of hyperprogressive disease (HPD) in patients receiving immune checkpoint inhibitors (ICIs). We compared the incidence of HPD during treatment with nivolumab±ipilimumab versus natural tumor progression with placebo in post hoc analyses of two randomized, double-blind clinical trials. METHODS: ATTRACTION-2 randomized patients with advanced gastric or gastroesophageal junction cancer (GC/GEJC) and progression on ≥2 prior regimens to nivolumab 3 mg/kg Q2W or placebo. CheckMate 451 randomized patients with extensive-disease small cell lung cancer (ED SCLC) and ongoing complete/partial response or stable disease after first-line chemotherapy to nivolumab 240 mg Q2W, nivolumab 1 mg/kg+ipilimumab 3 mg/kg Q3W for four doses then nivolumab 240 mg Q2W, or placebo. Patients receiving ≥1 dose of study drug and with tumor scans at baseline and the first on-treatment evaluation were included in the HPD analyses. HPD definitions were ≥20%, ≥50%, and ≥100% increase in target lesion sum of the longest diameters (SLD) at the first on-treatment assessment. RESULTS: In the ATTRACTION-2 HPD-evaluable population, 243 patients received nivolumab and 115 placebo. Fewer patients receiving nivolumab versus placebo had increases in SLD ≥20% (33.7% vs 46.1%) and ≥50% (6.2% vs 11.3%); similar proportions had increases in SLD ≥100% (1.6% vs 1.7%). In the CheckMate 451 HPD-evaluable population, 177 patients received nivolumab, 179 nivolumab+ipilimumab, and 175 placebo. Fewer patients receiving nivolumab or nivolumab+ipilimumab versus placebo had increases in SLD ≥20% (27.1%, 27.4% vs 45.7%), ≥50% (10.2%, 11.2% vs 22.3%), and ≥100% (2.8%, 2.8% vs 6.3%). CONCLUSIONS: Nivolumab±ipilimumab was not associated with an increased rate of progression versus placebo in patients with GC, GEJC, or ED SCLC, suggesting that previous reports of HPD may reflect the natural disease course in some patients rather than ICI-mediated progression. TRIAL REGISTRATION NUMBER: NCT02538666; NCT02267343.


Assuntos
Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Humanos , Ipilimumab/efeitos adversos , Neoplasias Pulmonares/patologia , Nivolumabe/efeitos adversos , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico
6.
Cancer Res ; 65(20): 9547-54, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16230420

RESUMO

T-cell adoptive immunotherapy for stringent murine tumor models, such as intracranial, s.c., or advanced pulmonary metastases, routinely uses lymphodepletive conditioning regimens before T-cell transfer, like recent clinical protocols. In this study, we examined whether host lymphodepletion is an obligatory component of curative T-cell therapy; we also examined the mechanism by which it augments therapy. Mice bearing intracranial, s.c., or 10-day pulmonary metastases of MCA 205 received total body irradiation conditioning or were nonirradiated before i.v. transfer of tumor-reactive T cells. Total body irradiation was not required for immunologically specific curative therapy and induction of memory provided that a 3- to 12-fold higher T-cell dose was administered. The mechanism involved enhanced intratumoral proliferation of T-effector cells in total body irradiation-conditioned recipients. In this tumor model, intratumoral T(reg) cells were not detected; consequently, intratumoral T-effector cells produced identical amounts of IFN-gamma upon ex vivo antigen stimulation irrespective of total body irradiation conditioning. Thus, host lymphodepletion augments T-cell immunotherapy through enhanced antigen-driven proliferation of T-effector cells, but curative therapy can be achieved in nonconditioned hosts by escalation of T-cell dose. These data provide a rationale for dose escalation of T-effector cells in situations where single or repeated lymphodepletion regimens are contraindicated.


Assuntos
Imunoterapia Adotiva/métodos , Linfócitos T/imunologia , Linfócitos T/efeitos da radiação , Irradiação Corporal Total , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Complexo CD3/imunologia , Terapia Combinada , Epitopos de Linfócito T/imunologia , Feminino , Fibrossarcoma/imunologia , Fibrossarcoma/secundário , Fibrossarcoma/terapia , Interleucina-2/imunologia , Interleucina-7/imunologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL
7.
J Neurosurg ; 103(1): 156-64, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16121986

RESUMO

OBJECT: Immunotherapy for malignant brain tumors by active immunization or adoptive transfer of tumor antigen-specific T lymphocytes has the potential to make up for some of the limitations of current clinical therapy. In this study, the authors tested whether active immunotherapy is curative in mice bearing advanced, rapidly progressive intracranial tumors. METHODS: Tumor vaccines were created through electrofusion of dendritic cells (DCs) and irradiated tumor cells to form multinucleated heterokaryons that retained the potent antigen processing and costimulatory function of DCs as well as the entire complement of tumor antigens. Murine hosts bearing intracranial GL261 glioma or MCA 205 fibrosarcoma were treated with a combination of local cranial radiotherapy, intrasplenic vaccination with DC/tumor fusion cells, and anti-OX40R (CD134) monoclonal antibody (mAb) 7 days after tumor inoculation. Whereas control mice had a median survival of approximately 20 days, the treated mice underwent complete tumor regression that was immunologically specific. Seven days after vaccination treated mice demonstrated robust infiltration of CD4+ and CD8+ T cells, which was exclusively confined to the tumor without apparent neurological toxicity. Cured mice survived longer than 120 days with no evidence of tumor recurrence and resisted intracranial tumor challenge. CONCLUSIONS: These data indicate a strategy to achieve an antitumor response against tumors in the central nervous system that is highly focused from both immunological and anatomical perspectives.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/fisiologia , Fibrossarcoma/tratamento farmacológico , Glioma/tratamento farmacológico , Imunoterapia Ativa/métodos , Animais , Neoplasias Encefálicas/radioterapia , Fusão Celular , Quimioterapia Adjuvante , Modelos Animais de Doenças , Feminino , Fibrossarcoma/radioterapia , Glioma/radioterapia , Camundongos , Camundongos Endogâmicos C57BL , Radioterapia Adjuvante , Receptores OX40 , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Receptores do Fator de Necrose Tumoral/uso terapêutico
8.
J Transl Med ; 2(1): 41, 2004 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-15566571

RESUMO

T cell-mediated cancer immunotherapy is dose dependent and optimally requires participation of antigen-specific CD4+ and CD8+ T cells. Here, we isolated tumor-sensitized T cells and activated them in vitro using conditions that led to greater than 108-fold numerical hyperexpansion of either the CD4+ or CD8+ subset while retaining their capacity for in vivo therapeutic efficacy. Murine tumor-draining lymph node (TDLN) cells were segregated to purify the CD62Llow subset, or the CD4+ subset thereof. Cells were then propagated through multiple cycles of anti-CD3 activation with IL-2 + IL-7 for the CD8+ subset, or IL-7 + IL-23 for the CD4+ subset. A broad repertoire of TCR Vbeta families was maintained throughout hyperexpansion, which was similar to the starting population. Adoptive transfer of hyper-expanded CD8+ T cells eliminated established pulmonary metastases, in an immunologically specific fashion without the requirement for adjunct IL-2. Hyper-expanded CD4+ T cells cured established tumors in intracranial or subcutaneous sites that were not susceptible to CD8+ T cells alone. Because accessibility and antigen presentation within metastases varies according to anatomic site, maintenance of a broad repertoire of both CD4+ and CD8+ T effector cells will augment the overall systemic efficacy of adoptive immunotherapy.

9.
Arch Immunol Ther Exp (Warsz) ; 51(4): 245-57, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12956434

RESUMO

The recognition by effector T lymphocytes of novel antigenic targets on tumor cells is the premise of specific, targeted immunotherapy of cancer. With the molecular characterization of peptide epitopes from melanoma antigens and, more recently, broadly expressed tumor antigens, there has been considerable enthusiasm for clinical evaluation of peptide tumor vaccines. Immunologic monitoring of vaccinated patients has demonstrated an expansion of CD8+ T cells that react with the relevant peptide and, more importantly, with native tumor. In most instances, however, vaccine-induced CD8+ T cell responses alone have not been sufficiently robust or sustained to translate into a high percentage of durable clinical responses. Vaccine strategies have also utilized dendritic cells (DCs) that have been modified to present tumor antigens. The superior antigen-processing capacity and co-stimulatory function of DCs convey a powerful stimulatory signal to both CD4+ and CD8+ T cells. Several strategies are attempting to broaden the immune response beyond single antigens by introducing the entire complement of tumor antigens into DCs. Adoptive immunotherapy is a promising strategy to recover tumor-reactive precursor T cells from patients, stimulate them to induce numerical expansion, and then re-infuse them. Ex vivo manipulation of the tumor-reactive T cells also permits cytotoxic therapy to be administered to the patient without damaging the effector cells. Recently, host lymphodepletion prior to adoptive transfer of effector T cells has resulted in an extremely high and sustained frequency of effectors that has achieved therapeutic efficacy against bulky metastatic disease in a substantial fraction of treated patients.


Assuntos
Vacinas Anticâncer , Imunoterapia Adotiva , Neoplasias/terapia , Linfócitos T/imunologia , Antígenos de Neoplasias/imunologia , Transplante de Células , Humanos , Imunoterapia Ativa , Ativação Linfocitária , Neoplasias/imunologia , Subpopulações de Linfócitos T/imunologia
10.
J Pediatr Surg ; 48(4): e1-3, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23583157

RESUMO

Osteosarcoma metastasis to the gastrointestinal tract is a rare phenomenon (Horiuchi A, Watanabe Y, Yoshida M, et al.: Metastatic osteosarcoma in the jejunum with intussusception: report of a case. Surg Today 2007;37:440-2). Gastrointestinal metastases may cause intussusception, bowel obstruction, or hemorrhage (Horiuchi A, Watanabe Y, Yoshida M, et al.: Metastatic osteosarcoma in the jejunum with intussusception: report of a case. Surg Today 2007;37:440-2; Chondramohan K, Somanathan T, Kusamakumary P: Metastatic osteosarcoma causing intussusception. J Pediatr Surg 2003;38(E44):1-3; Hung GY, Chiou TJ, Hsieh YL, et al.: Intestinal metastasis causing intussusception in a patient treated for osteosarcoma with history of multiple metastases: a case report. Jpn J Clin Oncol 2001;31:165-167). We report a case of a 17 year old male with osteosarcoma metastatic to the stomach and ascending colon, causing significant chronic gastrointestinal hemorrhage. Surgical resection was performed due to persistent, symptomatic anemia. The patient is free of recurrent hemorrhage at 24months after metastectomy. Resection of gastrointestinal metastases of osteosarcoma offers good palliation of chronic hemorrhage related to these lesions.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias do Colo/secundário , Neoplasias do Colo/cirurgia , Fíbula/patologia , Hemorragia Gastrointestinal/etiologia , Hemorragia Gastrointestinal/cirurgia , Osteossarcoma/secundário , Osteossarcoma/cirurgia , Neoplasias Gástricas/secundário , Neoplasias Gástricas/cirurgia , Adolescente , Biópsia , Diagnóstico Diferencial , Humanos , Masculino , Metastasectomia
11.
J Clin Anesth ; 25(4): 255-62, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23659826

RESUMO

STUDY OBJECTIVE: To assess preoperative and postoperative immune function in patients undergoing surgical resection of non-small cell lung cancer during general anesthesia and postoperative epidural analgesia. DESIGN: Observational single-center study. SETTING: University-affiliated academic center. PATIENTS: 24 adult, ASA physical status 3 and 4 patients with stage 1, 2, or 3 non-small cell lung cancer. No study patient received preoperative chemotherapy or radiation. INTERVENTIONS: Patients underwent thoracotomy with general anesthesia and postoperative epidural analgesia. MEASUREMENTS: Bispectral index monitoring, sevoflurane requirements, and intraoperative transfusions were recorded. Total fentanyl consumption and pain (verbal numeric rating scale) were recorded 24 hours after surgery. Preoperative and 24-hour postoperative natural killer cell percentage and function and percentages of natural killer T cells, T helper cells (CD4+), and cytotoxic T lymphocytes (CD8+) were measured. Plasma concentrations of the TH1 cytokine interleukin-2 and interferon-gamma and the TH2 cytokines interleukin-4 were measured at the same time points. RESULTS: The percentage (preoperative, 13.07 ± 9.81% vs postoperative, 9.6 ± 6.57%, P < 0.001) and function (preoperative, 31.61 ± 21.96%; postoperative, 13.61 ± 9.36%; P < 0.001) of natural killer cells was significantly decreased after surgery, but the percentage of natural killer T cells, T helper cells (CD4+), and cytotoxic T lymphocytes (CD8+) remained unchanged postoperatively; thus, the CD4/CD8 ratio remained unchanged. Postoperative plasma concentrations of the three cytokines were similar to preoperative levels; therefore, the TH1/TH2 ratio also remained unchanged. CONCLUSIONS: Innate immunity is depressed in patients with non-small cell lung cancer after surgical resection, and immunity is not preserved by the use of postoperative epidural analgesia.


Assuntos
Analgesia Epidural/métodos , Anestesia Geral/métodos , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Neoplasias Pulmonares/cirurgia , Idoso , Analgésicos Opioides/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/patologia , Monitores de Consciência , Citocinas/sangue , Feminino , Fentanila/administração & dosagem , Humanos , Imunidade Inata , Neoplasias Pulmonares/patologia , Masculino , Éteres Metílicos/administração & dosagem , Pessoa de Meia-Idade , Células T Matadoras Naturais/imunologia , Dor Pós-Operatória/tratamento farmacológico , Período Pós-Operatório , Sevoflurano
13.
Blood ; 109(11): 4865-76, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17284532

RESUMO

The importance of CD4+ Th1 cells during the effector phase of the antitumor response has been overshadowed by emphasis on CD8+ cytotoxic T lymphocytes (CTLs). To determine their respective functions, we purified antigen-primed T cells from tumor-draining lymph nodes and separately activated CD4+ and CD8+ subsets in vitro. Adoptive transfer of CD4+ T effector cells (T(E)s) combined with CD8+ T(E)s provided synergistic therapy for mice bearing subcutaneous, intracranial, or advanced pulmonary metastases. CD4+ T(E)s augmented IFN-gamma production by CD8+ T(E)s when cells were stimulated by tumor digest-containing antigen-presenting cells (APCs). CD4+ T(E)s infiltrated and proliferated extensively in pulmonary tumors, while also stimulating tumor antigen-specific CD8+ T cells. By contrast, CD8+ T(E)s showed minimal intratumoral proliferation in the absence of CD4+ cells or when systemically transferred CD4+ cells were prevented from infiltrating pulmonary tumors by pretreatment with pertussis toxin. Irradiation of CD4+ T cells immediately prior to adoptive transfer abrogated their intratumoral proliferation and direct antitumor efficacy but did not block their capacity to stimulate intratumoral CD8+ T(E) proliferation or tumor regression. These results highlight the importance of cross-presentation of tumor antigens during the effector phase of immunotherapy and suggest that approaches to stimulate CD4+ T(E) function and boost APC cross-presentation within tumors will augment cancer immunotherapy.


Assuntos
Antineoplásicos/farmacologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunoterapia/métodos , Neoplasias Pulmonares/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Proliferação de Células , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica
14.
Cell Immunol ; 243(1): 30-40, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17207783

RESUMO

Dendritic-tumor heterokaryons generated by electrofusion are highly immunogenic. In animal studies, a single vaccination was therapeutic for tumors established in the lung, skin, and brain. However, effective therapy required a third signal which could be provided by exogenous IL-12 or the agonistic anti-OX-40R monoclonal antibody (mAb). In this study, we investigated the mechanism and mode of actions of these two seemingly distinct adjuvants. In immunotherapy of the MCA205 sarcoma, administration of the neutralizing anti-IL-12 mAb nearly completely blocked the adjuvant effect of IL-12, but had minimal inhibitory effects on anti-OX-40R mAb. By contrast, in vivo administration of the antagonistic anti-OX-40L mAb inhibited the adjuvant effects of both IL-12 and anti-OX-40R mAb. Thus, a common pathway of endogenous OX-40 interaction is critical for the development of a therapeutic immune response. Analysis of the third signal mechanism revealed that in the absence of an adjuvant, vaccination with fusion hybrids led to IL-10 production without eliciting IFN-gamma secreting cells. The addition of IL-12 to vaccination suppressed IL-10 production and initiated sensitization of specific IFN-gamma secreting cells, resulting in a type 1-like antitumor immunity. These findings underscore the significance of the third signal in the design of dendritic cell-based cancer vaccines.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Imunidade Celular , Interleucina-12/farmacologia , Ligante OX40/farmacologia , Sarcoma Experimental/terapia , Animais , Anticorpos Monoclonais/imunologia , Fusão Celular , Células Dendríticas/metabolismo , Feminino , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-12/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Ligante OX40/uso terapêutico , Vacinação/métodos
15.
J Immunol ; 169(6): 3314-20, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12218152

RESUMO

Tumor-draining lymph nodes (TDLN) contain sensitized T cells with the phenotype CD62 L-selectin(low) (CD62L(low)) that can be activated ex vivo with anti-CD3 mAb and IL-2 to acquire potent dose-dependent effector function manifested upon adoptive transfer to secondary tumor-bearing hosts. In this study advanced tumor models were used as a stringent comparison of efficacy for the CD62L(low) subset, comprising 5-7% of the TDLN cells, vs the total population of TDLN cells following culture in high dose IL-2 (100 U/ml). During the 9-day activation period the total number of CD8+ T cells increased 1500-fold, with equivalent proliferation in the CD62L(low) vs the total TDLN cell cultures. Adoptive transfer of activated CD62L(low) cells eliminated 14-day pulmonary metastases and cured 10-day s.c. tumors, whereas transfer of maximally tolerated numbers of total TDLN cells was not therapeutic. Despite their propagation in a high concentration of IL-2, the hyperexpanded CD62L(low) subset of TDLN cells functioned in vivo without exogenous IL-2, and CD8+ T cells demonstrated relative helper independence. Moreover, the anti-tumor response was specific for the sensitizing tumor, and long term memory was established. The facile enrichment of tumor-reactive TDLN T cells, based on the CD62L(low) phenotype, circumvents the need for prior knowledge of the relevant tumor Ags. Coupling the isolation of pre-effector T cells with rapid ex vivo expansion to >3 logs could overcome some of the shortcomings of active immunotherapy or in vivo cytokine treatment, where selective robust expansion of effector cells has been difficult to achieve.


Assuntos
Imunoterapia Adotiva/métodos , Selectina L/biossíntese , Ativação Linfocitária , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Divisão Celular/imunologia , Citotoxicidade Imunológica , Feminino , Fibrossarcoma/induzido quimicamente , Fibrossarcoma/imunologia , Fibrossarcoma/patologia , Fibrossarcoma/terapia , Antígenos de Histocompatibilidade/imunologia , Memória Imunológica , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Linfonodos/imunologia , Linfonodos/patologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Indução de Remissão , Sarcoma Experimental/induzido quimicamente , Sarcoma Experimental/patologia , Subpopulações de Linfócitos T/patologia , Subpopulações de Linfócitos T/transplante , Células Tumorais Cultivadas
16.
J Immunol ; 172(6): 3462-8, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15004146

RESUMO

Adoptive transfer of tumor-specific effector T cells induces regression of advanced tumors and induces a long term memory response; however, the origin of this response has not been clearly defined. In this study Thy1.2+ mice bearing advanced MCA-205 tumors were treated with sublethal total body irradiation, followed by adoptive transfer of congenic Thy1.1+ T cells that had been sensitized to tumor in vivo and then activated ex vivo with anti-CD3, IL-2, and IL-7. Splenocytes were recovered >140 days after the initial therapy, and the L-selectinlow memory cell subset was separated into host Thy1.2+ and transferred Thy1.1+ cells and restimulated ex vivo. Both adoptively transferred Thy1.1+ cells as well as reconstituted host Thy1.2+ cells could specifically eliminate MCA-205 pulmonary metastases. Interestingly, hosts with partial responses followed by tumor recurrence nevertheless harbored memory cells that could be isolated and numerically amplified ex vivo to regenerate potent effector function. Memory cells were recovered after adoptive transfer into lymphodepleted nontumor-bearing hosts, indicating that they were not dependent on continued Ag exposure. These experiments establish that rapid ex vivo expansion of tumor Ag-primed T cells does not abrogate their capacity to become long-lived memory cells. Moreover, immune-mediated tumor regression coincident with lymphoid reconstitution produces another wave of host memory cells. These data suggest an approach to rescuing antitumor immune function even in hosts with long-standing progressive tumor through restorative ex vivo activation.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica , Imunoterapia Adotiva , Depleção Linfocítica , Sarcoma Experimental/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/transplante , Linfócitos T Reguladores/transplante , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Feminino , Imunoterapia Adotiva/métodos , Injeções Subcutâneas , Depleção Linfocítica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias/patologia , Sarcoma Experimental/patologia , Sarcoma Experimental/prevenção & controle , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Antígenos Thy-1/biossíntese
17.
J Immunol ; 169(9): 4811-21, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391191

RESUMO

Tumor-specific effector T cells (T(E)) are naturally sensitized within the L-selectin(low) (CD62L(low)) fraction of tumor-draining lymph nodes (TDLN). Whether isolated from day 9 (D9) or day 12 (D12) TDLN, 5 million L-selectin(low) T(E) could be culture activated and adoptively transferred to achieve complete rejection of established intradermal, pulmonary, and brain tumors. Surprisingly, although 25 million unfractionated T cells from D9 TDLN were equally effective, even 100 million unfractionated T cells from D12 TDLN seldom prevented lethal intradermal tumor progression, despite a pronounced therapeutic excess of T(E). This highly reproducible treatment failure was due to cotransfer of tumor-induced, L-selectin(high) suppressor T cells (T(S)) which were also present in D12 TDLN. In contrast, D9 TDLN and normal spleens lacked L-selectin(high) T(S). Only those L-selectin(high) D12 TDLN T cells that down-regulated L-selectin during culture activation were suppressive in vivo and in vitro, and, like L-selectin(low) T(E), trafficked promptly into tumors following i.v. administration. This is the first demonstration that adoptive immunotherapy can fail as a direct result of passenger T(S) that share certain phenotypic and trafficking features of T(E), even when otherwise curative doses of T(E) have been administered. Furthermore, in contrast to recently described CD4(+)CD25(+) T(S) and plasmacytoid dendritic cell-activated T(S), tumor-induced L-selectin(high) T(S) prevent tumor rejection via blockade of sensitized, activated T(E) rather than via afferent blockade.


Assuntos
Antígenos de Neoplasias/imunologia , Fibrossarcoma/imunologia , Fibrossarcoma/terapia , Imunoterapia Adotiva/métodos , Selectina L/biossíntese , Linfócitos T Reguladores/imunologia , Animais , Movimento Celular/imunologia , Separação Celular , Progressão da Doença , Regulação para Baixo/imunologia , Feminino , Fibrossarcoma/patologia , Antígenos de Histocompatibilidade/imunologia , Injeções Intradérmicas , Interferon gama/antagonistas & inibidores , Interferon gama/biossíntese , Selectina L/fisiologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/transplante , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Linfócitos do Interstício Tumoral/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/transplante , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante , Falha de Tratamento , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA