Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
EMBO J ; 40(11): e106658, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33855732

RESUMEN

Cytotoxic T cells (CTLs) can eliminate tumor cells through the delivery of lethal hits, but the actual efficiency of this process in the tumor microenvironment is unclear. Here, we visualized the capacity of single CTLs to attack tumor cells in vitro and in vivo using genetically encoded reporters that monitor cell damage and apoptosis. Using two distinct malignant B-cell lines, we found that the majority of cytotoxic hits delivered by CTLs in vitro were sublethal despite proper immunological synapse formation, and associated with reversible calcium elevation and membrane damage in the targets. Through intravital imaging in the bone marrow, we established that the majority of CTL interactions with lymphoma B cells were either unproductive or sublethal. Functional heterogeneity of CTLs contributed to diverse outcomes during CTL-tumor contacts in vivo. In the therapeutic settings of anti-CD19 CAR T cells, the majority of CAR T cell-tumor interactions were also not associated with lethal hit delivery. Thus, differences in CTL lytic potential together with tumor cell resistance to cytotoxic hits represent two important bottlenecks for anti-tumor responses in vivo.


Asunto(s)
Inmunoterapia Adoptiva , Linfoma/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos CD19/inmunología , Apoptosis , Linfocitos B/inmunología , Calcio/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Sinapsis Inmunológicas/inmunología , Linfoma/terapia , Ratones , Ratones Endogámicos C57BL , Receptores Quiméricos de Antígenos/inmunología
3.
Trends Immunol ; 40(11): 1022-1034, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31668676

RESUMEN

Understanding complex interactions between the immune system and the tumor microenvironment is an essential step towards the rational development and optimization of immunotherapies. Several experimental approaches are available to tackle this complexity but most are not designed to address the dynamic features of immune reactions, including cell migration, cellular interactions, and transient signaling events. By providing a unique means to access these precious parameters, intravital imaging offers a fresh look at intratumoral immune responses at the single-cell level. Here, we discuss how in vivo imaging sheds light on fundamental aspects of tumor immunity and helps elucidate modes of action of immunotherapies. We conclude by discussing future developments that may consolidate the unique contribution of intravital imaging for our understanding of tumor immunity.


Asunto(s)
Inmunoterapia/métodos , Microscopía Intravital/métodos , Neoplasias/inmunología , Animales , Movimiento Celular , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Vigilancia Inmunológica , Monitorización Inmunológica , Neoplasias/terapia , Escape del Tumor , Microambiente Tumoral
4.
Br J Haematol ; 193(4): 814-826, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33844842

RESUMEN

Pure red cell aplasia (PRCA) following allogeneic haematopoietic stem cell transplantation (aHSCT) with major ABO incompatibility is responsible for transfusion dependent anaemia, impaired quality of life and iron overload. We conducted a retrospective study, over a 10-year period, which included all consecutive patients who received a major ABO mismatched aHSCT, to assess the impact of specific treatment on PRCA. We did not observe any PRCA in the 57 aHSCT issued from cord blood. Among the remaining 631 patients, cumulative incidence of PRCA was 10·5% [range 8·2-13.0]. The median duration of resolved PRCA was 171 days [IQR 116; 261]. Pre-transplant high isohaemagglutinins titre was associated with an increased risk of PRCA (P < 10-4 ). PRCA did not affect overall survival (P = 0·95). Twenty-two patients (33·3%) received at least one specific treatment. The most commonly used treatments were rituximab (17 patients) and donor lymphocyte infusion (DLI; seven patients). Regarding PRCA resolution, we did not observe a significant difference between treated or untreated subjects (HR = 0·93, 95% confidence interval (CI) 0·48- 1·80; P = 0·82). Similar results were observed with erythropoietin treatment (22 patients, HR = 0·86 95% CI: [0·47-1·57] P = 0·62). Our data do not support the use of erythropoietin, rituximab or DLI for the treatment of PRCA.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/sangre , Incompatibilidad de Grupos Sanguíneos , Trasplante de Células Madre Hematopoyéticas , Aplasia Pura de Células Rojas , Adolescente , Adulto , Anciano , Aloinjertos , Incompatibilidad de Grupos Sanguíneos/sangre , Incompatibilidad de Grupos Sanguíneos/mortalidad , Niño , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Aplasia Pura de Células Rojas/sangre , Aplasia Pura de Células Rojas/mortalidad , Aplasia Pura de Células Rojas/terapia , Tasa de Supervivencia
6.
Leukemia ; 38(2): 326-339, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38148396

RESUMEN

Current recommended risk scores to predict thrombotic events associated with myeloproliferative neoplasms (MPN) do not discriminate between arterial and venous thrombosis despite their different physiopathology. To define novel stratification systems, we delineated a comprehensive landscape of MPN associated thrombosis across a large long-term follow-up MPN cohort. Prior arterial thrombosis, age >60 years, cardiovascular risk factors and presence of TET2 or DNMT3A mutations were independently associated with arterial thrombosis in multivariable analysis. ARTS, an ARterial Thrombosis Score, based on these four factors, defined low- (0.37% patients-year) and high-risk (1.19% patients-year) patients. ARTS performance was superior to the two-tiered conventional risk stratification in our training cohort, across all MPN subtypes, as well as in two external validation cohorts. Prior venous thrombosis and presence of a JAK2V617F mutation with a variant allelic frequency ≥50% were independently associated with venous thrombosis. The discrimination potential of VETS, a VEnous Thrombosis Score based on these two factors, was poor, similar to the two-tiered conventional risk stratification. Our study pinpoints arterial and venous thrombosis clinico-molecular differences and proposes an arterial risk score for more accurate patients' stratification. Further improvement of venous risk scores, accounting for additional factors and considering venous thrombosis as a heterogeneous entity is warranted.


Asunto(s)
Trastornos Mieloproliferativos , Neoplasias , Trombosis , Trombosis de la Vena , Humanos , Persona de Mediana Edad , Neoplasias/complicaciones , Trombosis de la Vena/genética , Trombosis/genética , Trombosis/complicaciones , Mutación , Trastornos Mieloproliferativos/complicaciones , Trastornos Mieloproliferativos/genética , Factores de Riesgo , Janus Quinasa 2/genética , Medición de Riesgo
7.
Cell Rep Med ; 4(9): 101161, 2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37595589

RESUMEN

Anti-CD19 chimeric antigen receptor (CAR) T cell therapy represents a breakthrough for the treatment of B cell malignancies. Yet, it can lead to severe adverse events, including cytokine release syndrome (CRS), which may require urgent clinical management. Whether interpatient variability in CAR T cell subsets contributes to CRS is unclear. Here, we show that CD4+ CAR T cells are the main drivers of CRS. Using an immunocompetent model of anti-CD19 CAR T cell therapy, we report that CD4+, but not CD8+, CAR T cells elicit physiological CRS-like manifestations associated with the release of inflammatory cytokines. In CAR T cell-treated patients, CRS occurrence and severity are significantly associated with high absolute values of CD4+ CAR T cells in the blood. CRS in mice occurs independently of CAR T cell-derived interferon γ (IFN-γ) but requires elevated tumor burden. Thus, adjusting the CD4:CD8 CAR T cell ratio to patient tumor load may help mitigate CAR T cell-associated toxicities.


Asunto(s)
Síndrome de Liberación de Citoquinas , Inmunoterapia Adoptiva , Humanos , Animales , Ratones , Síndrome de Liberación de Citoquinas/etiología , Inmunoterapia Adoptiva/efectos adversos , Linfocitos T CD8-positivos , Antígenos CD19 , Linfocitos T CD4-Positivos
8.
Nat Cancer ; 4(7): 968-983, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37248395

RESUMEN

CD4+ T cells and CD4+ chimeric antigen receptor (CAR) T cells display highly variable antitumor activity in preclinical models and in patients; however, the mechanisms dictating how and when CD4+ T cells promote tumor regression are incompletely understood. With the help of functional intravital imaging, we report that interferon (IFN)-γ production but not perforin-mediated cytotoxicity was the dominant mechanism for tumor elimination by anti-CD19 CD4+ CAR T cells. Mechanistically, mouse or human CD4+ CAR T-cell-derived IFN-γ diffused extensively to act on tumor cells at distance selectively killing tumors sensitive to cytokine-induced apoptosis, including antigen-negative variants. In anti-CD19 CAR T-cell-treated patients exhibiting elevated CAR CD4:CD8 ratios, strong induction of serum IFN-γ was associated with increased survival. We propose that the sensitivity of tumor cells to the pro-apoptotic activity of IFN-γ is a major determinant of CD4+ CAR T-cell efficacy and may be considered to guide the use of CD4+ T cells during immunotherapy.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Animales , Ratones , Receptores de Antígenos de Linfocitos T , Citocinas , Interferón gamma , Linfocitos T CD4-Positivos
9.
Nat Commun ; 13(1): 3111, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35661707

RESUMEN

Cytotoxic T cells are important components of natural anti-tumor immunity and are harnessed in tumor immunotherapies. Immune responses to tumors and immune therapy outcomes largely vary among individuals, but very few studies examine the contribution of intrinsic behavior of the T cells to this heterogeneity. Here we show the development of a microfluidic-based in vitro method to track the outcome of antigen-specific T cell activity on many individual cancer spheroids simultaneously at high spatiotemporal resolution, which we call Multiscale Immuno-Oncology on-Chip System (MIOCS). By combining parallel measurements of T cell behaviors and tumor fates with probabilistic modeling, we establish that the first recruited T cells initiate a positive feedback loop to accelerate further recruitment to the spheroid. We also provide evidence that cooperation between T cells on the spheroid during the killing phase facilitates tumor destruction. Thus, we propose that both T cell accumulation and killing function rely on collective behaviors rather than simply reflecting the sum of individual T cell activities, and the possibility to track many replicates of immune cell-tumor interactions with the level of detail our system provides may contribute to our understanding of immune response heterogeneity.


Asunto(s)
Microfluídica , Neoplasias , Humanos , Inmunoterapia , Neoplasias/patología , Linfocitos T Citotóxicos , Microambiente Tumoral
10.
Sci Immunol ; 6(57)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33771887

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy relies on the activity of a large pool of tumor-targeting cytotoxic effectors. Whether CAR T cells act autonomously or require interactions with the tumor microenvironment (TME) remains incompletely understood. Here, we report an essential cross-talk between CAR T cell subsets and the TME for tumor control in an immunocompetent mouse B cell lymphoma model of anti-CD19 CAR T cell therapy. Using single-cell RNA sequencing, we revealed substantial modification of the TME during CAR T cell therapy. Interferon-γ (IFN-γ) produced by CAR T cells not only enhanced endogenous T and natural killer cell activity but was also essential for sustaining CAR T cell cytotoxicity, as revealed by intravital imaging. CAR T cell-derived IFN-γ facilitated host interleukin-12 production that supported host immune and CAR T cell responses. Compared with CD8+ CAR T cells, CD4+ CAR T cells were more efficient at host immune activation but less capable of direct tumor killing. In summary, CAR T cells do not act independently in vivo but rely instead on cytokine-mediated cross-talk with the TME for optimal activity. Invigorating CAR T cell interplay with the host represents an attractive strategy to prevent relapses after therapy.


Asunto(s)
Comunicación Celular/inmunología , Pruebas Inmunológicas de Citotoxicidad , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Microambiente Tumoral/inmunología , Animales , Antígenos CD19/inmunología , Antígenos de Neoplasias/inmunología , Comunicación Celular/genética , Línea Celular Tumoral , Biología Computacional/métodos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inmunoterapia Adoptiva , Interferón gamma/biosíntesis , Activación de Linfocitos/inmunología , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Linfoma de Células B/terapia , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética
11.
Nat Cancer ; 1(3): 302-314, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32803171

RESUMEN

The cytokine IFN-γ produced by tumor-reactive T cells is a key effector molecule with pleiotropic effects during anti-tumor immune responses. While IFN-γ production is targeted at the immunological synapse, its spatiotemporal activity within the tumor remains elusive. Here, we report that while IFN-γ secretion requires local antigen recognition, IFN-γ diffuses extensively to alter the tumor microenvironment in distant areas. Using intravital imaging and a reporter for STAT1 translocation, we provide evidence that T cells mediate sustained IFN-γ signaling in remote tumor cells. Furthermore, tumor phenotypic alterations required several hours of exposure to IFN-γ, a feature that disfavored local IFN-γ activity over diffusion and bystander activity. Finally, single-cell RNA-seq data from melanoma patients also suggested bystander IFN-γ activity in human tumors. Thus, tumor-reactive T cells act collectively to create large cytokine fields that profoundly modify the tumor microenvironment.


Asunto(s)
Interferón gamma , Microambiente Tumoral , Citocinas , Humanos , Linfocitos T
12.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA