Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 67(10): 1519-1531, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30039427

RESUMEN

Epithelial ovarian cancer (EOC) is the leading cause of gynaecological cancer-related death in Europe. Although most patients achieve an initial complete response with first-line treatment, recurrence occurs in more than 80% of cases. Thus, there is a clear unmet need for novel second-line treatments. EOC is frequently infiltrated with T lymphocytes, the presence of which has been shown to be associated with improved clinical outcomes. Adoptive T-cell therapy (ACT) using ex vivo-expanded tumour-infiltrating lymphocytes (TILs) has shown remarkable efficacy in other immunogenic tumours, and may represent a promising therapeutic strategy for EOC. In this preclinical study, we investigated the efficacy of using anti-CD3/anti-CD28 magnetic beads and IL-2 to expand TILs from freshly resected ovarian tumours. TILs were expanded for up to 3 weeks, and then subjected to a rapid-expansion protocol (REP) using irradiated feeder cells. Tumours were collected from 45 patients with EOC and TILs were successfully expanded from 89.7% of biopsies. Expanded CD4+ and CD8+ subsets demonstrated features associated with memory phenotypes, and had significantly higher expression of key activation and functional markers than unexpanded TILs. Expanded TILs produced anti-tumour cytokines when co-cultured with autologous tumour cells, inferring tumour cytotoxicity. Our findings demonstrate that it is possible to re-activate and expand tumour-reactive T cells from ovarian tumours. This presents a promising immunotherapy that could be used sequentially or in combination with current therapeutic strategies.


Asunto(s)
Adenocarcinoma de Células Claras/terapia , Carcinosarcoma/terapia , Cistadenocarcinoma Seroso/terapia , Citocinas/metabolismo , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Ováricas/terapia , Adenocarcinoma de Células Claras/inmunología , Adenocarcinoma de Células Claras/metabolismo , Anciano , Carcinosarcoma/inmunología , Carcinosarcoma/metabolismo , Cistadenocarcinoma Seroso/inmunología , Cistadenocarcinoma Seroso/metabolismo , Citotoxicidad Inmunológica , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Células Tumorales Cultivadas
2.
Immunol Rev ; 257(1): 91-106, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24329792

RESUMEN

Blockbuster antibody therapies have catapulted immune-based approaches to treat cancer into the consciousness of mainstay clinical research. On the back of this, other emerging immune-based therapies are providing great promise. T-cell therapy is one such area where recent trials using T cells genetically modified to express an antibody-based chimeric antigen receptor (CAR) targeted against the CD19 antigen have demonstrated impressive responses when adoptively transferred to patients with advanced chronic lymphocytic leukemia. The general concept of the CAR T cell was devised some 20 years ago. In this relatively short period of time, the technology to redirect T-cell function has moved at pace facilitating clinical translation; however, many questions remain with respect to developing the approach to improve CAR T-cell therapeutic activity and also to broaden the range of tumors that can be effectively targeted by this approach. This review highlights some of the underlying principles and compromises of CAR T-cell technology using the CD19-targeted CAR as a paradigm and discusses some of the issues that relate to targeting solid tumors with CAR T cells.


Asunto(s)
Antígenos CD19/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Técnicas de Cultivo de Célula , Técnicas de Transferencia de Gen , Ingeniería Genética , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Leucemia de Células B/inmunología , Leucemia de Células B/terapia , Linfoma de Células B/inmunología , Linfoma de Células B/terapia
3.
J Immunol ; 192(8): 3654-65, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24623129

RESUMEN

T cells engrafted with chimeric AgRs (CAR) are showing exciting potential for targeting B cell malignancies in early-phase clinical trials. To determine whether the second-generation CAR was essential for optimal antitumor activity, two CD28-based CAR constructs targeting CD19 were tested for their ability to redirect mouse T cell function against established B cell lymphoma in a BALB/c syngeneic model system. T cells armed with either CAR eliminated A20 B cell lymphoma in vivo; however, one construct induced a T cell dose-dependent acute toxicity associated with a raised serum Th1 type cytokine profile on transfer into preconditioned mice. Moreover, a chronic toxicity manifested as granuloma-like formation in spleen, liver, and lymph nodes was observed in animals receiving T cells bearing either CD28 CAR, albeit with different kinetics dependent upon the specific receptor used. This phenotype was associated with an expansion of CD4+ CAR+ T cells and CD11b+ Gr-1(+) myeloid cells and increased serum Th2-type cytokines, including IL-10 and IL-13. Mouse T cells engrafted with a first-generation CAR failed to develop such autotoxicity, whereas toxicity was not apparent when T cells bearing the same receptors were transferred into C57BL/6 or C3H animals. In summary, the adoptive transfer of second-generation CD19-specific CAR T cells can result in a cell dose-dependent acute toxicity, whereas the prolonged secretion of high levels of Th2 cytokines from these CAR T cells in vivo drives a granulomatous reaction resulting in chronic toxicity. Strategies that prevent a prolonged Th2-cytokine biased CAR T cell response are clearly warranted.


Asunto(s)
Antígenos CD19/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Traslado Adoptivo/efectos adversos , Animales , Antígenos CD19/genética , Análisis por Conglomerados , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Linfoma de Células B/terapia , Ratones , Ratones Endogámicos BALB C , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Subgrupos de Linfocitos T/trasplante , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
4.
Cancer Immunol Immunother ; 63(2): 133-45, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24190544

RESUMEN

Adoptive cell therapy employing gene-modified T-cells expressing chimeric antigen receptors (CARs) has shown promising preclinical activity in a range of model systems and is now being tested in the clinical setting. The manufacture of CAR T-cells requires compliance with national and European regulations for the production of medicinal products. We established such a compliant process to produce T-cells armed with a first-generation CAR specific for carcinoembryonic antigen (CEA). CAR T-cells were successfully generated for 14 patients with advanced CEA(+) malignancy. Of note, in the majority of patients, the defined procedure generated predominantly CD4(+) CAR T-cells with the general T-cell population bearing an effector-memory phenotype and high in vitro effector function. Thus, improving the process to generate less-differentiated T-cells would be more desirable in the future for effective adoptive gene-modified T-cell therapy. However, these results confirm that CAR T-cells can be generated in a manner compliant with regulations governing medicinal products in the European Union.


Asunto(s)
Traslado Adoptivo , Antígeno Carcinoembrionario/inmunología , Proteínas Quimerinas/biosíntesis , Receptores de Antígenos de Linfocitos T/biosíntesis , Linfocitos T/inmunología , Humanos , Inmunofenotipificación , Interferón gamma/biosíntesis
5.
Blood ; 119(15): 3523-33, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22354003

RESUMEN

Monoclonal antibodies (mAbs) have revolutionized the treatment of B-cell malignancies. Although Fc-dependent mechanisms of mAb-mediated tumor clearance have been extensively studied, the ability of mAbs to directly evoke programmed cell death (PCD) in the target cell and the underlying mechanisms involved remain under-investigated. We recently demonstrated that certain mAbs (type II anti-CD20 and anti-HLA DR mAbs) potently evoked PCD through an actin-dependent, lysosome-mediated process. Here, we reveal that the induction of PCD by these mAbs, including the type II anti-CD20 mAb GA101 (obinutuzumab), directly correlates with their ability to produce reactive oxygen species (ROS) in human B-lymphoma cell lines and primary B-cell chronic lymphocytic leukemia cells. ROS scavengers abrogated mAb-induced PCD indicating that ROS are required for the execution of cell death. ROS were generated downstream of mAb-induced actin cytoskeletal reorganization and lysosome membrane permeabilization. ROS production was independent of mitochondria and unaffected by BCL-2 overexpression. Instead, ROS generation was mediated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. These findings provide further insights into a previously unrecognized role for NADPH oxidase-derived ROS in mediating nonapoptotic PCD evoked by mAbs in B-cell malignancies. This newly characterized cell death pathway may potentially be exploited to eliminate malignant cells, which are refractory to conventional chemotherapy and immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Leucemia/patología , Linfoma/patología , Especies Reactivas de Oxígeno/farmacología , Actinas/metabolismo , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD20/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia/genética , Leucemia/metabolismo , Linfoma/genética , Linfoma/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/fisiología , NADPH Oxidasa 2 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , NADPH Oxidasas/fisiología , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
6.
Blood ; 117(17): 4519-29, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21378274

RESUMEN

The anti-CD20 mAb rituximab has substantially improved the clinical outcome of patients with a wide range of B-cell malignancies. However, many patients relapse or fail to respond to rituximab, and thus there is intense investigation into the development of novel anti-CD20 mAbs with improved therapeutic efficacy. Although Fc-FcγR interactions appear to underlie much of the therapeutic success with rituximab, certain type II anti-CD20 mAbs efficiently induce programmed cell death (PCD), whereas rituximab-like type I anti-CD20 mAbs do not. Here, we show that the humanized, glycoengineered anti-CD20 mAb GA101 and derivatives harboring non-glycoengineered Fc regions are type II mAb that trigger nonapoptotic PCD in a range of B-lymphoma cell lines and primary B-cell malignancies. We demonstrate that GA101-induced cell death is dependent on actin reorganization, can be abrogated by inhibitors of actin polymerization, and is independent of BCL-2 overexpression and caspase activation. GA101-induced PCD is executed by lysosomes which disperse their contents into the cytoplasm and surrounding environment. Taken together, these findings reveal that GA101 is able to potently elicit actin-dependent, lysosomal cell death, which may potentially lead to improved clearance of B-cell malignancies in vivo.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Linfoma de Células B , Actinas/efectos de los fármacos , Actinas/inmunología , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino/farmacología , Catepsinas/farmacología , Adhesión Celular/inmunología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/inmunología , Resistencia a Antineoplásicos/inmunología , Humanos , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Lisosomas/efectos de los fármacos , Lisosomas/inmunología , Rituximab
7.
Oncoimmunology ; 12(1): 2223094, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37332616

RESUMEN

Despite breakthroughs in immune checkpoint inhibitors (ICI), the majority of tumors, including those poorly infiltrated by CD8+ T cells or heavily infiltrated by immunosuppressive immune effector cells, are unlikely to result in clinically meaningful tumor responses. Radiation therapy (RT) has been combined with ICI to potentially overcome this resistance and improve response rates but reported clinical trial results have thus far been disappointing. Novel approaches are required to overcome this resistance and reprogram the immunosuppressive tumor microenvironment (TME) and address this major unmet clinical need. Using diverse preclinical tumor models of prostate and bladder cancer, including an autochthonous prostate tumor (Pten-/-/trp53-/-) that respond poorly to radiation therapy (RT) and anti-PD-L1 combinations, the key drivers of this resistance within the TME were profiled and used to develop rationalized combination therapies that simultaneously enhance activation of anti-cancer T cell responses and reprogram the immunosuppressive TME. The addition of anti-CD40mAb to RT resulted in an increase in IFN-y signaling, activation of Th-1 pathways with an increased infiltration of CD8+ T-cells and regulatory T-cells with associated activation of the CTLA-4 signaling pathway in the TME. Anti-CTLA-4mAb in combination with RT further reprogrammed the immunosuppressive TME, resulting in durable, long-term tumor control. Our data provide novel insights into the underlying mechanisms of the immunosuppressive TME that result in resistance to RT and anti-PD-1 inhibitors and inform therapeutic approaches to reprogramming the immune contexture in the TME to potentially improve tumor responses and clinical outcomes.


Asunto(s)
Microambiente Tumoral , Neoplasias de la Vejiga Urinaria , Masculino , Humanos , Linfocitos T Reguladores/metabolismo , Transducción de Señal , Terapia Combinada , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/radioterapia
8.
Front Cell Dev Biol ; 11: 1270398, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38020903

RESUMEN

Introduction: The glycoengineered type II anti-CD20 monoclonal antibody obinutuzumab has been licensed for treatment in follicular non-Hodgkin lymphoma and B-CLL following clinical trials demonstrating superior outcomes to standard of care treatment. However, ultimately many patients still relapse, highlighting the need to understand the mechanisms behind treatment failure to improve patient care. Resistance to chemotherapy is often caused by the ability of malignant B-cells to migrate to the bone marrow and home into the stromal layer. Therefore, this study aimed to investigate whether stromal cells were also able to inhibit type II anti-CD20 antibody mechanisms of action, contributing to resistance to therapy. Methods: A stromal-tumor co-culture was established in vitro between Raji or Daudi B-cell tumor cells and M210B4 stromal cells in 24 well plates. Results: Contact with stromal cells was able to protect tumor cells from obinutuzumab mediated programmed cell death (PCD), antibody dependent cellular phagocytosis and antibody dependent cellular cytotoxicity. Furthermore, such protection required direct contact between stroma and tumor cells. Stromal cells appeared to interfere with obinutuzumab mediated B-cell homotypic adhesion through inhibiting and reversing actin remodelling, potentially as a result of stromal-tumor cell contact leading to downregulation of CD20 on the surface of tumor cells. Further evidence for the potential role of CD20 downregulation comes through the reduction in surface CD20 expression and inhibition of obinutuzumab mediated PCD when tumor cells are treated with Ibrutinib in the presence of stromal cells. The proteomic analysis of tumor cells after contact with stromal cells led to the identification of a number of altered pathways including those involved in cell adhesion and the actin cytoskeleton and remodeling. Discussion: This work demonstrates that contact between tumor cells and stromal cells leads to inhibition of Obinutuzumab effector functions and has important implications for future therapies to improve outcomes to anti-CD20 antibodies. A deeper understanding of how anti-CD20 antibodies interact with stromal cells could prove a useful tool to define better strategies to target the micro-environment and ultimately improve patient outcomes in B-cell malignancies.

9.
J Immunol ; 184(4): 1885-96, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20089697

RESUMEN

T cells gene-modified to express chimeric Ag receptors (CARs) have shown potent antitumor activity in vivo and are in clinical trials at locations worldwide. However, CAR activity has been investigated in mouse models in which Ag expression is restricted to the tumor. To explore the impact of normal tissue expression of the target Ag, we developed a mouse CD19-specific CAR to investigate antitumor efficacy against a syngeneic B cell lymphoma cell line within a background of normal CD19(+) host B cells. Mouse T cells engrafted with the amCD19CD3zeta CAR specifically lysed A20 lymphoma targets and B cells in vitro. These T cells also eradicated a 12-d established disseminated A20 lymphoma in mice preconditioned with 6 Gy total body irradiation. In the short-term (7 d after adoptive transfer), amCD19z T cells underwent Ag-dependent proliferation in vivo with a concomitant depletion in host B cell levels. However, the levels of amCD19z CAR(+) T cells decreased significantly at later time points, at which point host B cells returned, eventually reaching normal levels. In contrast, CAR(+) T cells lacking a signaling domain or specificity for mCD19 persisted over extended periods in blood and spleen. Importantly, no overt clinical signs of autotoxicity were observed in tumor-free or tumor-bearing mice treated with amCD19z T cells over an extended period of time. These observations highlight the importance of studying the activity of CAR(+) T cells in autologous models that have the normal range of tissue expression of Ag.


Asunto(s)
Traslado Adoptivo , Antígenos CD19/biosíntesis , Antígenos CD19/genética , Antineoplásicos/farmacología , Epítopos de Linfocito T/inmunología , Marcación de Gen , Linfocitos T/inmunología , Linfocitos T/metabolismo , Traslado Adoptivo/métodos , Animales , Antígenos CD19/inmunología , Antineoplásicos/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Marcación de Gen/métodos , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/genética , Retroviridae/genética , Retroviridae/inmunología , Linfocitos T/trasplante , Acondicionamiento Pretrasplante
10.
J Clin Oncol ; 40(20): 2203-2212, 2022 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-35385334

RESUMEN

PURPOSE: There is a need to refine the selection of patients with oropharyngeal squamous cell carcinoma (OPSCC) for treatment de-escalation. We investigated whether pretreatment absolute lymphocyte count (ALC) predicted overall survival (OS) benefit from the addition of concurrent chemotherapy to radical radiotherapy. PATIENTS AND METHODS: This was an observational study of consecutive OPSCCs treated by curative-intent radiotherapy, with or without concurrent chemotherapy (n = 791) with external, independent validation from a separate institution (n = 609). The primary end point was OS at 5 years. Locoregional control (LRC) was assessed using competing risk regression as a secondary end point. Previously determined prognostic factors were used in a multivariable Cox proportional hazards model to assess the prognostic importance of ALC and the interaction between ALC and cisplatin chemotherapy use. RESULTS: Pretreatment ALC was prognostic for 5-year OS on multivariable analysis (hazard ratio [HR] 0.64; 95% CI, 0.42 to 0.98; P = .04). It also predicted benefit from the use of concurrent cisplatin chemotherapy, with a significant interaction between cisplatin chemotherapy and pretreatment ALC (likelihood ratio test, P = .04): higher ALC count reduced the 5-year OS benefit compared with radiotherapy alone (HR 2.53; 95% CI, 1.03 to 6.19; P = .043). This was likely driven by an effect on LRC up to 5 years (interaction subdistribution HR 2.29; 95% CI, 0.68 to 7.71; P = .094). An independent validation cohort replicated the OS (HR 2.53; 95% CI, 0.98 to 6.52; P = .055) and LRC findings (interaction subdistribution HR 3.43; 95% CI, 1.23 to 9.52; P = .018). CONCLUSION: For OPSCC, the pretreatment ALC is prognostic for OS and also predicts benefit from the addition of cisplatin chemotherapy to radiotherapy. These findings require prospective evaluation, and could inform the selection of good prognosis patients for a de-escalation trial.


Asunto(s)
Cisplatino , Neoplasias Orofaríngeas , Supervivencia sin Enfermedad , Humanos , Recuento de Linfocitos , Neoplasias Orofaríngeas/tratamiento farmacológico , Neoplasias Orofaríngeas/radioterapia , Pronóstico , Modelos de Riesgos Proporcionales
11.
Cancers (Basel) ; 12(10)2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-33008040

RESUMEN

Radiotherapy (RT) is a highly effective anti-cancer treatment. Immunotherapy using immune checkpoint blockade (ICI) has emerged as a new and robust pillar in cancer therapy; however, the response rate to single agent ICI is low whilst toxicity remains. Radiotherapy has been shown to have local and systemic immunomodulatory effects. Therefore, combining RT and immunotherapy is a rational approach to enhance anti-tumour immune responses. However, the immunomodulatory effects of RT can be both immunostimulatory or immunosuppressive and may be different across different tumour types and patients. Therefore, there is an urgent medical need to establish biomarkers to guide clinical decision making in predicting responses or in patient selection for RT-based combination treatments. In this review, we summarize the immunological effects of RT on the tumour microenvironment and emerging biomarkers to help better understand the implications of these immunological changes, and we provide new insights into the potential for combination therapies with RT and immunotherapy.

12.
Radiat Oncol ; 15(1): 254, 2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33148287

RESUMEN

Radiotherapy (RT) is a highly effective anti-cancer therapy delivered to around 50-60% of patients. It is part of therapy for around 40% of cancer patients who are cured of their disease. Until recently, the focus of this anti-tumour efficacy has been on the direct tumour cytotoxicity and RT-induced DNA damage. Recently, the immunomodulatory effects of RT on the tumour microenvironment have increasingly been recognized. There is now intense interest in potentially using RT to induce an anti-tumour immune response, which has led to rethinking into how the efficacy of RT could be further enhanced. Following the breakthrough of immune check point inhibitors (ICIs), a new era of immuno-oncology (IO) agents has emerged and established immunotherapy as a routine part of cancer treatment. Despite ICI improving outcomes in many cancer types, overall durable responses occur in only a minority of patients. The immunostimulatory effects of RT make combinations with ICI attractive to potentially amplify anti-tumour immunity resulting in increased tumour responses and improved outcomes. In contrast, tumours with profoundly immunosuppressive tumour microenvironments, dominated by myeloid-derived cell populations, remain a greater clinical challenge and RT may potentially further enhance the immunosuppression. To harness the full potential of RT and IO agent combinations, further insights are required to enhance our understanding of the role these immunosuppressive myeloid populations play, how RT influences these populations and how they may be therapeutically manipulated in combination with RT to improve outcomes further. These are exciting times with increasing numbers of IO targets being discovered and IO agents undergoing clinical evaluation. Multidisciplinary research collaborations will be required to establish the optimal parameters for delivering RT (target volume, dose and fractionation) in combination with IO agents, including scheduling to achieve maximal therapeutic efficacy.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/radioterapia , Microambiente Tumoral , Terapia Combinada , Humanos , Inmunoterapia , Neoplasias/inmunología , Linfocitos T/fisiología
13.
J Exp Med ; 216(5): 1038-1049, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30936262

RESUMEN

CAR T cells represent a potentially curative strategy for B cell malignancies. However, the outcome and dynamics of CAR T cell interactions in distinct anatomical sites are poorly understood. Using intravital imaging, we tracked interactions established by anti-CD19 CAR T cells in B cell lymphoma-bearing mice. Circulating targets trapped CAR T cells in the lungs, reducing their access to lymphoid organs. In the bone marrow, tumor apoptosis was largely due to CAR T cells that engaged, killed, and detached from their targets within 25 min. Notably, not all CAR T cell contacts elicited calcium signaling or killing while interacting with tumors, uncovering extensive functional heterogeneity. Mathematical modeling revealed that direct killing was sufficient for tumor regression. Finally, antigen-loss variants emerged in the bone marrow, but not in lymph nodes, where CAR T cell cytotoxic activity was reduced. Our results identify a previously unappreciated level of diversity in the outcomes of CAR T cell interactions in vivo, with important clinical implications.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Microscopía Intravital/métodos , Linfoma de Células B/terapia , Receptores Quiméricos de Antígenos/metabolismo , Análisis de la Célula Individual/métodos , Linfocitos T/metabolismo , Animales , Antígenos CD19/metabolismo , Apoptosis , Línea Celular Tumoral , Pulmón/metabolismo , Linfoma de Células B/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Modelos Teóricos , Recurrencia
14.
Br J Haematol ; 142(1): 65-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18477047

RESUMEN

T cells genetically engineered to express tumour-targeting receptors are attractive anti-cancer therapeutic agents. Human T cells engrafted with a chimeric receptor specific for the B-cell lymphoma antigen CD19 fused to the CD3zeta receptor (aCD19z) are functional in vitro. Current successful clinical protocols targeting melanoma use pre-conditioning chemotherapy in combination with T cells. This study demonstrated that interleukin-2 expanded aCD19z T cells combined with cyclophosphamide effectively treated five-day established Raji B-cell lymphoma in an immunocompromised model system with 50% of mice surviving >100 days. This observation strongly supports the combination of antibody targeted T cells with chemotherapy as a novel approach for the therapy of CD19(+) B-cell malignancies.


Asunto(s)
Antígenos CD19/administración & dosificación , Ciclofosfamida/administración & dosificación , Inmunoconjugados/administración & dosificación , Linfoma de Células B/terapia , Linfocitos T , Animales , Ingeniería Genética , Vectores Genéticos , Ratones , Ratones SCID
16.
Clin Cancer Res ; 13(19): 5719-28, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17908961

RESUMEN

PURPOSE: To study the production of chemokines by colorectal hepatic metastases. EXPERIMENTAL DESIGN: Biopsies of resected colorectal hepatic metastases and nonneoplastic adjacent liver tissue were screened for chemokines using protein arrays and results were confirmed by ELISA and immunohistochemistry. RESULTS: Two chemokines, eotaxin-2 and MCP-1, were found at elevated levels within the tumor biopsy compared with adjacent liver. The relative increase in expression from tumor was much higher for eotaxin-2 than MCP-1, with 10 of 25 donors having a >100-fold increase in expression compared with 0 of 24 donors for MCP-1. In a parallel analysis, eotaxin-2 was also found at elevated levels in the tumor region of primary colorectal cancer biopsies. Immunohistochemical staining indicated that carcinoembryonic antigen-positive tumor cells stained strongly for eotaxin-2, implicating these cells as the predominant source of the chemokine. In vitro studies confirmed that several colorectal tumor lines produce eotaxin-2 and that secretion of this chemokine could be depressed by IFN-gamma and enhanced by the Th2-type cytokines interleukin-4 and interleukin-13. Jurkat T cells were engineered to express the receptor for eotaxin-2 (CCR3). These cells effectively migrated in response to eotaxin-2 protein, suggesting that immune cells gene modified to express a chemokine receptor may have improved abilities to home to tumor. CONCLUSIONS: Taken together, these observations confirm eotaxin-2 as a chemokine strongly associated with primary and metastatic tumors of colorectal origin. Furthermore, the importance of this result may be a useful tool in the development of targeted therapeutic approaches to colorectal tumors.


Asunto(s)
Quimiocina CCL24/fisiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/terapia , Regulación Neoplásica de la Expresión Génica , Inmunoterapia/métodos , Neoplasias Hepáticas/secundario , Antineoplásicos/uso terapéutico , Biopsia , Línea Celular Tumoral , Movimiento Celular , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Neoplasias Colorrectales/inmunología , Humanos , Sistema Inmunológico , Metástasis de la Neoplasia
17.
J Immunother ; 41(3): 130-140, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29239915

RESUMEN

Chimeric antigen receptor (CAR) T cells represent a novel targeted approach to overcome both quantitative and qualitative shortfalls of the host immune system relating to the detection and subsequent destruction of tumors. The identification of antigens expressed specifically on the surface of tumor cells is a critical first step in the ability to utilize CAR T cells for the treatment of cancer. The 5T4 is a tumor-associated antigen which is expressed on the cell surface of most solid tumors including ovarian cancer. Matched blood and tumor samples were collected from 12 patients with ovarian cancer; all tumors were positive for 5T4 expression by immunohistochemistry. Patient T cells were effectively transduced with 2 different anti-5T4 CAR constructs which differed in their affinity for the target antigen. Co-culture of CAR T cells with matched autologous tumor disaggregates resulted in antigen-specific secretion of IFN-gamma. Furthermore, assessment of the efficacy of anti-5T4 CAR T cells in a mouse model resulted in therapeutic benefit against established ovarian tumors. These results demonstrate proof of principle that 5T4 is an attractive target for immune intervention in ovarian cancer and that patient T cells engineered to express a 5T4-specific CAR can recognize and respond physiologically to autologous tumor cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia Adoptiva , Glicoproteínas de Membrana/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Inmunoterapia Adoptiva/métodos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptores Quiméricos de Antígenos/genética , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Clin Cancer Res ; 23(18): 5514-5526, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28533222

RESUMEN

Purpose: Radiotherapy is a highly effective anticancer treatment forming part of the standard of care for the majority of patients, but local and distal disease recurrence remains a major cause of mortality. Radiotherapy is known to enhance tumor immunogenicity; however, the contribution and mechanisms of radiotherapy-induced immune responses are unknown.Experimental Design: The impact of low-dose fractionated radiotherapy (5 × 2 Gy) alone and in combination with αPD-1 mAb on the tumor microenvironment was evaluated by flow cytometry and next-generation sequencing of the T-cell receptor (TCR) repertoire. A dual-tumor model was used, with fractionated radiotherapy delivered to a single tumor site to enable evaluation of the local and systemic response to treatment and ability to induce abscopal responses outside the radiation field.Results: We show that fractionated radiotherapy leads to T-cell infiltration at the irradiated site; however, the TCR landscape remains dominated by polyclonal expansion of preexisting T-cell clones. Adaptive resistance via the PD-1/PD-L1 pathway restricts the generation of systemic anticancer immunity following radiotherapy, which can be overcome through combination with αPD-1 mAb leading to improved local and distal tumor control. Moreover, we show that effective clearance of tumor following combination therapy is dependent on both T cells resident in the tumor at the time of radiotherapy and infiltrating T cells.Conclusions: These data provide evidence that radiotherapy can enhance T-cell trafficking to locally treated tumor sites and augment preexisting anticancer T-cell responses with the capacity to mediate regression of out-of-field tumor lesions when delivered in combination with αPD-1 mAb therapy. Clin Cancer Res; 23(18); 5514-26. ©2017 AACR.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Neoplasias/inmunología , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/efectos de la radiación , Animales , Línea Celular Tumoral , Terapia Combinada , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Neoplasias/metabolismo , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/metabolismo , Radioterapia , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Tasa de Supervivencia , Subgrupos de Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Curr Opin Mol Ther ; 8(1): 62-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16506527

RESUMEN

Micromet AG and Medlmmune Inc are developing MT-103, a single-chain bispecific recombinant antibody from Micromet's BiTE (bispecific T-cell engager) product platform that binds both the CD19 antigen and the T-cell receptor (CD3), for the potential treatment of B-cell lymphoma. The company is also investigating the compound for the potential treatment of chronic lymphocytic leukemia and acute lymphoblastic leukemia.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Industria Farmacéutica , Animales , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/farmacocinética , Ensayos Clínicos Fase I como Asunto , Humanos , Linfoma de Células B/tratamiento farmacológico , Relación Estructura-Actividad
20.
Leuk Lymphoma ; 57(6): 1269-80, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27050042

RESUMEN

Nearly two decades ago rituximab heralded a new era in management of B cell malignancies significantly increasing response rates and survival. However, despite clear therapeutic advantage, significant numbers of patients become refractory to anti-CD20 mAb therapy, suggesting urgent improvements are required. It is now well recognized that the suppressive tumor microenvironment plays an important role in the outcome of anti-CD20 mAb therapy and that manipulation of this environment may improve the efficacy and produce long-term tumor control. The past few years have seen a surge of interest in immunomodulatory agents capable of overwriting immune suppressive networks into favorable clinical outcome. Currently, a number of such combinations with anti-CD20 mAb is under evaluation and some have produced encouraging outcomes in rituximab refractory disease. In this review, we give an outline of anti-CD20 mAbs and explore the combinations with immunomodulatory agents that enhance antitumor immunity through targeting stimulatory or inhibitory pathways and have proven potential to synergize with anti-CD20 mAb therapy. These agents, primarily mAbs, target CTLA-4, PD-1/PD-L1, and CD40.


Asunto(s)
Antineoplásicos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Inmunomodulación/efectos de los fármacos , Leucemia de Células B/tratamiento farmacológico , Linfoma de Células B/tratamiento farmacológico , Rituximab/uso terapéutico , Animales , Antígenos CD20 , Antineoplásicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígenos CD40/antagonistas & inhibidores , Antígeno CTLA-4/antagonistas & inhibidores , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Factores Inmunológicos/farmacología , Leucemia de Células B/inmunología , Leucemia de Células B/metabolismo , Leucemia de Células B/patología , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Terapia Molecular Dirigida , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Rituximab/farmacología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA