Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Clin Invest ; 132(17)2022 09 01.
Article de Anglais | MEDLINE | ID: mdl-35881486

RÉSUMÉ

The in vivo persistence of adoptively transferred T cells is predictive of antitumor response. Identifying functional properties of infused T cells that lead to in vivo persistence and tumor eradication has remained elusive. We profiled CD19-specific chimeric antigen receptor (CAR) T cells as the infusion products used to treat large B cell lymphomas using high-throughput single-cell technologies based on time-lapse imaging microscopy in nanowell grids (TIMING), which integrates killing, cytokine secretion, and transcriptional profiling. Our results show that the directional migration of CD19-specific CAR T cells is correlated with multifunctionality. We showed that CD2 on T cells is associated with directional migration and that the interaction between CD2 on T cells and CD58 on lymphoma cells accelerates killing and serial killing. Consistent with this, we observed that elevated CD58 expression on pretreatment tumor samples in patients with relapsed or refractory large B cell lymphomas treated with CD19-specific CAR T cell therapy was associated with complete clinical response and survival. These results highlight the importance of studying dynamic T cell-tumor cell interactions in identifying optimal antitumor responses.


Sujet(s)
Antigènes CD2/métabolisme , Antigènes CD58/métabolisme , Lymphome B diffus à grandes cellules , Lymphocytes T , Antigènes CD19 , Humains , Immunothérapie adoptive/méthodes , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/thérapie , Récepteurs aux antigènes des cellules T , Analyse sur cellule unique
2.
Biotechniques ; 72(4): 113-120, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35297693

RÉSUMÉ

Understanding immune response to infections and vaccines lags understanding humoral responses. While neutralizing antibody responses wane over time, T cells are instrumental in long-term immunity. We apply machine learning and time-lapse imaging microscopy in nanowell grids (TIMING) to study thousands of videos of T cells with specificity for SARS-CoV-2 eliminating targets bearing spike protein as a surrogate for viral infection. The data on effector functions, including cytokine secretion and cytotoxicity, provide the first direct evidence that cytotoxic T lymphocytes from a convalescent patient targeting an epitope conserved across all known variants of concern are serial killers capable of eliminating multiple infected target cells. These data have implications for vaccine development and for the recovery and monitoring of infected individuals.


Sujet(s)
COVID-19 , SARS-CoV-2 , Anticorps antiviraux , Vaccins contre la COVID-19 , Épitopes , Humains , Glycoprotéine de spicule des coronavirus , Lymphocytes T cytotoxiques
4.
J Immunother Cancer ; 9(3)2021 03.
Article de Anglais | MEDLINE | ID: mdl-33722906

RÉSUMÉ

BACKGROUND: Adoptive cell therapy based on the infusion of chimeric antigen receptor (CAR) T cells has shown remarkable efficacy for the treatment of hematologic malignancies. The primary mechanism of action of these infused T cells is the direct killing of tumor cells expressing the cognate antigen. However, understanding why only some T cells are capable of killing, and identifying mechanisms that can improve killing has remained elusive. METHODS: To identify molecular and cellular mechanisms that can improve T-cell killing, we utilized integrated high-throughput single-cell functional profiling by microscopy, followed by robotic retrieval and transcriptional profiling. RESULTS: With the aid of mathematical modeling we demonstrate that non-killer CAR T cells comprise a heterogeneous population that arise from failure in each of the discrete steps leading to the killing. Differential transcriptional single-cell profiling of killers and non-killers identified CD137 as an inducible costimulatory molecule upregulated on killer T cells. Our single-cell profiling results directly demonstrate that inducible CD137 is feature of killer (and serial killer) T cells and this marks a different subset compared with the CD107apos (degranulating) subset of CAR T cells. Ligation of the induced CD137 with CD137 ligand (CD137L) leads to younger CD19 CAR T cells with sustained killing and lower exhaustion. We genetically modified CAR T cells to co-express CD137L, in trans, and this lead to a profound improvement in anti-tumor efficacy in leukemia and refractory ovarian cancer models in mice. CONCLUSIONS: Broadly, our results illustrate that while non-killer T cells are reflective of population heterogeneity, integrated single-cell profiling can enable identification of mechanisms that can enhance the function/proliferation of killer T cells leading to direct anti-tumor benefit.


Sujet(s)
Ligand de 4-1BB/génétique , Analyse de profil d'expression de gènes , Immunothérapie adoptive , Leucémies/thérapie , Tumeurs de l'ovaire/thérapie , Récepteurs chimériques pour l'antigène/génétique , Analyse sur cellule unique , Lymphocytes T/transplantation , Transcriptome , Ligand de 4-1BB/métabolisme , Animaux , Cytotoxicité immunologique/génétique , Femelle , Récepteur cellulaire-2 du virus de l'hépatite A/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Humains , Immunophénotypage , Cellules K562 , Leucémies/génétique , Leucémies/immunologie , Leucémies/métabolisme , Souris de lignée NOD , Souris SCID , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/métabolisme , Phénotype , Récepteurs chimériques pour l'antigène/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Oncoimmunology ; 8(10): e1051298, 2019.
Article de Anglais | MEDLINE | ID: mdl-31646063

RÉSUMÉ

Genetically engineered T cells that express chimeric antigen receptors (CAR+) are heterogeneous and thus, understanding the immunotherapeutic efficacy remains a challenge in adoptive cell therapy. We developed a high-throughput single-cell methodology, Timelapse Imaging Microscopy In Nanowell Grids (TIMING) to monitor interactions between immune cells and tumor cells in vitro. Using TIMING we demonstrated that CD4+ CAR+ T cells participate in multi-killing and benefit from improved resistance to activation induced cell death in comparison to CD8+ CAR+ T cells. For both subsets of cells, effector cell fate at the single-cell level was dependent on functional activation through multiple tumor cells.

7.
JCI Insight ; 1(17): e87754, 2016 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-27777972

RÉSUMÉ

Current antiangiogenesis therapy relies on inhibiting newly developed immature tumor blood vessels and starving tumor cells. This strategy has shown transient and modest efficacy. Here, we report a better approach to target cancer-associated endothelial cells (ECs), reverse permeability and leakiness of tumor blood vessels, and improve delivery of chemotherapeutic agents to the tumor. First, we identified deregulated microRNAs (miRs) from patient-derived cancer-associated ECs. Silencing these miRs led to decreased vascular permeability and increased maturation of blood vessels. Next, we screened a thioaptamer (TA) library to identify TAs selective for tumor-associated ECs. An annexin A2-targeted TA was identified and used for delivery of miR106b-5p and miR30c-5p inhibitors, resulting in vascular maturation and antitumor effects without inducing hypoxia. These findings could have implications for improving vascular-targeted therapy.


Sujet(s)
Antinéoplasiques/pharmacologie , Aptamères nucléotidiques , Cellules endothéliales/cytologie , microARN/administration et posologie , Néovascularisation pathologique/prévention et contrôle , Lignée cellulaire tumorale , Humains , Nanoparticules , Tumeurs/vascularisation , Tumeurs/thérapie , Transfection
8.
Mol Ther ; 24(7): 1178-86, 2016 08.
Article de Anglais | MEDLINE | ID: mdl-27203439

RÉSUMÉ

Chimeric antigen receptor (CAR) endows specificity to T-cells independent of human leukocyte antigen (HLA). This enables one immunoreceptor to directly target the same surface antigen on different subsets of tumor cells from multiple HLA-disparate recipients. Most approaches manufacture individualized CAR(+)T-cells from the recipient or HLA-compatible donor, which are revealing promising clinical results. This is the impetus to broaden the number of patients eligible to benefit from adoptive immunotherapy such as to infuse third-party donor derived CAR(+)T-cells. This will overcome issues associated with (i) time to manufacture T-cells, (ii) cost to generate one product for one patient, (iii) inability to generate a product from lymphopenic patients or patient's immune cells fail to complete the manufacturing process, and (iv) heterogeneity of T-cell products produced for or from individual recipients. Establishing a biobank of allogeneic genetically modified immune cells from healthy third-party donors, which are cryopreserved and validated in advance of administration, will facilitate the centralizing manufacturing and widespread distribution of CAR(+)T-cells to multiple points-of-care in a timely manner. To achieve this, it is necessary to engineer an effective strategy to avoid deleterious allogeneic immune responses leading to toxicity and rejection. We review the strategies to establish "off-the-shelf" donor-derived biobanks for human application of CAR(+)T-cells as a drug.


Sujet(s)
Expression des gènes , Immunothérapie adoptive , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie , Protéines de fusion recombinantes , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , , Animaux , Thérapie cellulaire et tissulaire , Thérapie génétique , Maladie du greffon contre l'hôte/étiologie , Maladie du greffon contre l'hôte/prévention et contrôle , Humains , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Tumeurs/immunologie , Tumeurs/thérapie
9.
Mol Ther ; 24(6): 1078-1089, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-26945006

RÉSUMÉ

Neoantigens unique to each patient's tumor can be recognized by autologous T cells through their T-cell receptor (TCR) but the low frequency and/or terminal differentiation of mutation-specific T cells in tumors can limit their utility as adoptive T-cell therapies. Transfer of TCR genes into younger T cells from peripheral blood with a high proliferative potential could obviate this problem. We generated a rapid, cost-effective strategy to genetically engineer cancer patient T cells with TCRs using the clinical Sleeping Beauty transposon/transposase system. Patient-specific TCRs reactive against HLA-A*0201-restriced neoantigens AHNAK(S2580F) or ERBB2(H473Y) or the HLA-DQB*0601-restricted neoantigen ERBB2IP(E805G) were assembled with murine constant chains and cloned into Sleeping Beauty transposons. Patient peripheral blood lymphocytes were coelectroporated with SB11 transposase and Sleeping Beauty transposon, and transposed T cells were enriched by sorting on murine TCRß (mTCRß) expression. Rapid expansion of mTCRß(+) T cells with irradiated allogeneic peripheral blood lymphocytes feeders, OKT3, interleukin-2 (IL-2), IL-15, and IL-21 resulted in a preponderance of effector (CD27(-)CD45RA(-)) and less-differentiated (CD27(+)CD45RA(+)) T cells. Transposed T cells specifically mounted a polyfunctional response against cognate mutated neoantigens and tumor cell lines. Thus, Sleeping Beauty transposition of mutation-specific TCRs can facilitate the use of personalized T-cell therapy targeting unique neoantigens.


Sujet(s)
Tumeurs/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Lymphocytes T/anatomopathologie , Transposases/métabolisme , Animaux , Éléments transposables d'ADN , Génie génétique , Antigène HLA-A2/métabolisme , Chaines bêta des antigènes HLA-DQ/métabolisme , Humains , Protéines membranaires/immunologie , Souris , Récepteur ErbB-2/immunologie , Lymphocytes T/immunologie
10.
Oncoimmunology ; 4(11): e1047582, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26451325

RÉSUMÉ

We have previously reported that human endogenous retrovirus-K (HERV-K) envelope (env) protein is a tumor-associated antigen (TAA) for cancer vaccines, and that its antibodies (mAbs) possess antitumor activity against cancer. In this study, a chimeric antigen receptor (CAR) specific for HERV-K env protein (K-CAR) was generated using anti-HERV-K mAb. K-CAR T cells from peripheral blood mononuclear cells (PBMCs) of 9 breast cancer (BC) patients and 12 normal donors were able to inhibit growth of, and to exhibit significant cytotoxicity toward, BC cells but not MCF-10A normal breast cells. The antitumor effects in cancer cells were significantly reduced when control T cells were used, or the expression of HERV-K was knocked down by an shRNA. Secretion of multiple cytokines, including IFNγ, TNF-α, and IL-2, was significantly enhanced in culture media of BC cells treated with K-CARs. Significantly reduced tumor growth and tumor weight was observed in xenograft models bearing MDA-MB-231 or MDA-MB-435.eB1 BC cells. Importantly, the K-CAR prevented tumor metastasis to other organs. Furthermore, downregulation of HERV-K expression in tumors of mice treated with K-CAR correlated with upregulation of p53 and downregulation of MDM2 and p-ERK. Importantly, the expression of HERV-K env protein in metastatic tumor tissues treated with K-CAR T cells correlated with the expression of Ras. Our results indicate that HERV-K env protein is an oncoprotein and may play an important role in tumorigenesis related to p53 and Ras signaling pathways. Anti-HERV-K treatment, including K-CAR treatment, shows potential for immunotherapy of BC.

11.
Cancer Res ; 72(1): 271-81, 2012 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-22075555

RÉSUMÉ

The treatment of osteosarcoma pulmonary metastases remains a challenge. T cells genetically modified to express a chimeric antigen receptor (CAR), which recognizes a tumor-associated antigen, have shown activity against hematopoietic malignancies in clinical trials, but this requires the identification of a specific receptor on the tumor cell. In the current study, we found that interleukin (IL)-11Rα was selectively expressed on 14 of 16 osteosarcoma patients' lung metastases and four different human osteosarcoma cell lines, indicating that IL-11Rα may be a novel target for CAR-specific T-cell therapy. IL-11Rα expression was absent or low in normal organ tissues, with the exception of the gastrointestinal tract. IL-11Rα-CAR-specific T cells were obtained by non-viral gene transfer of Sleeping Beauty DNA plasmids and selectively expanded ex vivo using artificial antigen-presenting cells derived from IL-11Rα + K562 cells genetically modified to coexpress T-cell costimulatory molecules. IL-11Rα-CAR(+) T cells killed all four osteosarcoma cell lines in vitro; cytotoxicity correlated with the level of IL-11Rα expression on the tumor cells. Intravenous injection of IL-11Rα-CAR(+) T cells into mice resulted in the regression of osteosarcoma pulmonary metastases with no organ toxicity. Together, the data suggest that IL-11Rα-CAR T cells may represent a new therapy for patients with osteosarcoma pulmonary metastases.


Sujet(s)
Tumeurs osseuses/anatomopathologie , Sous-unité alpha du récepteur à l'interleukine-11/métabolisme , Tumeurs du poumon/secondaire , Ostéosarcome/anatomopathologie , Lymphocytes T/métabolisme , Animaux , Lignée cellulaire tumorale , Électroporation , Cytométrie en flux , Humains , Immunohistochimie , Souris , Souris nude , Métastase tumorale , Plasmides
13.
Mol Ther ; 17(4): 742-9, 2009 Apr.
Article de Anglais | MEDLINE | ID: mdl-19209146

RÉSUMÉ

Adoptive T-cell immunotherapy has shown promise in the treatment of human malignancies, but the challenge of isolating T cells with high avidity for tumor antigens in each patient has limited application of this approach. The transfer into T cells of T-cell receptor (TCR) genes encoding high-affinity TCRs recognizing defined tumor-associated antigens can potentially circumvent this obstacle. Using a well-characterized murine model of adoptive T-cell immunotherapy for widely disseminated leukemia, we demonstrate that TCR gene-modified T cells can cure mice of disseminated tumor. One goal of such adoptive therapy is to establish a persistent memory response to prevent recurrence; however, long-term function of transferred TCR-transduced T cells is limited due to reduced expression of the introduced TCR in vivo in quiescent resting T cells. However, by introducing the TCR into a cell with a known endogenous specificity, activation of these T cells by stimulation through the endogenous TCR can be used to increase expression of the introduced TCR, potentially providing a strategy to increase the total number of tumor-reactive T cells in the host and restore more potent antitumor activity.


Sujet(s)
Lymphocytes T CD8+/immunologie , Immunothérapie adoptive , Leucémie expérimentale/thérapie , Récepteurs aux antigènes des cellules T/génétique , Animaux , Vecteurs génétiques , Leucémie expérimentale/immunologie , Souris , Souris de lignée C57BL , Transduction génétique , Transfection
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...