Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
2.
J Vis Exp ; (203)2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38314829

RESUMEN

Glioblastoma, IDH-wild type, CNS WHO grade 4 (GBM) is a primary brain tumor associated with poor patient survival despite aggressive treatment. Developing realistic ex vivo models remain challenging. Patient-derived 3-dimensional organoid (PDO) models offer innovative platforms that capture the phenotypic and molecular heterogeneity of GBM, while preserving key characteristics of the original tumors. However, manual dissection for PDO generation is time-consuming, expensive and can result in a number of irregular and unevenly sized PDOs. This study presents an innovative method for PDO production using an automated tissue chopper. Tumor samples from four GBM and one astrocytoma, IDH-mutant, CNS WHO grade 2 patients were processed manually as well as using the tissue chopper. In the manual approach, the tumor material was dissected using scalpels under microscopic control, while the tissue chopper was employed at three different angles. Following culture on an orbital shaker at 37 °C, morphological changes were evaluated using bright field microscopy, while proliferation (Ki67) and apoptosis (CC3) were assessed by immunofluorescence after 6 weeks. The tissue chopper method reduced almost 70% of the manufacturing time and resulted in a significantly higher PDOs mean count compared to the manually processed tissue from the second week onwards (week 2: 801 vs. 601, P = 0.018; week 3: 1105 vs. 771, P = 0.032; and week 4:1195 vs. 784, P < 0.01). Quality assessment revealed similar rates of tumor-cell apoptosis and proliferation for both manufacturing methods. Therefore, the automated tissue chopper method offers a more efficient approach in terms of time and PDO yield. This method holds promise for drug- or immunotherapy-screening of GBM patients.


Asunto(s)
Astrocitoma , Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Neoplasias Encefálicas/patología , Glioma/patología , Glioblastoma/patología , Astrocitoma/patología , Organoides/patología
3.
Nat Commun ; 15(1): 993, 2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38307904

RESUMEN

The concept of precision cell therapy targeting tumor-specific mutations is appealing but requires surface-exposed neoepitopes, which is a rarity in cancer. B cell receptors (BCR) of mature lymphoid malignancies are exceptional in that they harbor tumor-specific-stereotyped sequences in the form of point mutations that drive self-engagement of the BCR and autologous signaling. Here, we use a BCR light chain neoepitope defined by a characteristic point mutation (IGLV3-21R110) for selective targeting of a poor-risk subset of chronic lymphocytic leukemia (CLL) with chimeric antigen receptor (CAR) T cells. We develop murine and humanized CAR constructs expressed in T cells from healthy donors and CLL patients that eradicate IGLV3-21R110 expressing cell lines and primary CLL cells, but neither cells expressing the non-pathogenic IGLV3-21G110 light chain nor polyclonal healthy B cells. In vivo experiments confirm epitope-selective cytolysis in xenograft models in female mice using engrafted IGLV3-21R110 expressing cell lines or primary CLL cells. We further demonstrate in two humanized mouse models lack of cytotoxicity towards human B cells. These data provide the basis for advanced approaches of resistance-preventive and biomarker-guided cellular targeting of functionally relevant lymphoma driver mutations sparing normal B cells.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Humanos , Femenino , Ratones , Animales , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/terapia , Linfocitos B , Mutación , Receptores de Antígenos de Linfocitos B/genética , Linfocitos T
4.
Cancers (Basel) ; 15(10)2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37345035

RESUMEN

While glioblastoma (GBM) is still challenging to treat, novel immunotherapeutic approaches have shown promising effects in preclinical settings. However, their clinical breakthrough is hampered by complex interactions of GBM with the tumor microenvironment (TME). Here, we present an analysis of TME composition in a patient-derived organoid model (PDO) as well as in organotypic slice cultures (OSC). To obtain a more realistic model for immunotherapeutic testing, we introduce an enhanced PDO model. We manufactured PDOs and OSCs from fresh tissue of GBM patients and analyzed the TME. Enhanced PDOs (ePDOs) were obtained via co-culture with PBMCs (peripheral blood mononuclear cells) and compared to normal PDOs (nPDOs) and PT (primary tissue). At first, we showed that TME was not sustained in PDOs after a short time of culture. In contrast, TME was largely maintained in OSCs. Unfortunately, OSCs can only be cultured for up to 9 days. Thus, we enhanced the TME in PDOs by co-culturing PDOs and PBMCs from healthy donors. These cellular TME patterns could be preserved until day 21. The ePDO approach could mirror the interaction of GBM, TME and immunotherapeutic agents and may consequently represent a realistic model for individual immunotherapeutic drug testing in the future.

5.
Sci Transl Med ; 14(664): eabh1209, 2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36170447

RESUMEN

Aspergillus fumigatus is a ubiquitous mold that can cause severe infections in immunocompromised patients, typically manifesting as invasive pulmonary aspergillosis (IPA). Adaptive and innate immune cells that respond to A. fumigatus are present in the endogenous repertoire of patients with IPA but are infrequent and cannot be consistently isolated and expanded for adoptive immunotherapy. Therefore, we gene-engineered A. fumigatus-specific chimeric antigen receptor (Af-CAR) T cells and demonstrate their ability to confer antifungal reactivity in preclinical models in vitro and in vivo. We generated a CAR targeting domain AB90-E8 that recognizes a conserved protein antigen in the cell wall of A. fumigatus hyphae. T cells expressing the Af-CAR recognized A. fumigatus strains and clinical isolates and exerted a direct antifungal effect against A. fumigatus hyphae. In particular, CD8+ Af-CAR T cells released perforin and granzyme B and damaged A. fumigatus hyphae. CD8+ and CD4+ Af-CAR T cells produced cytokines that activated macrophages to potentiate the antifungal effect. In an in vivo model of IPA in immunodeficient mice, CD8+ Af-CAR T cells localized to the site of infection, engaged innate immune cells, and reduced fungal burden in the lung. Adoptive transfer of CD8+ Af-CAR T cells conferred greater antifungal efficacy compared to CD4+ Af-CAR T cells and an improvement in overall survival. Together, our study illustrates the potential of gene-engineered T cells to treat aggressive infectious diseases that are difficult to control with conventional antimicrobial therapy and support the clinical development of Af-CAR T cell therapy to treat IPA.


Asunto(s)
Aspergilosis Pulmonar Invasiva , Receptores Quiméricos de Antígenos , Animales , Antifúngicos , Aspergillus fumigatus , Citocinas , Granzimas , Aspergilosis Pulmonar Invasiva/terapia , Ratones , Perforina , Linfocitos T
6.
Oncogene ; 41(34): 4104-4114, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35859167

RESUMEN

The success of chimeric antigen receptor T cell (CAR-T) therapy in the treatment of hematologic malignancies has prompted the development of numerous CAR-T technologies, including switchable CAR-T (sCAR-T) systems that combine a universal CAR-T with bispecific adapter proteins. Owing to their controllability and versatility, sCAR-Ts have received considerable attention. To explore the therapeutic utility of sCAR-Ts targeting the receptor tyrosine kinase ROR1, which is expressed in hematologic and solid malignancies, and to identify bispecific adaptor proteins that efficiently mediate universal CAR-T engagement, a panel of switches based on ROR1-targeting Fabs with different epitopes and affinities was compared in in vitro and in vivo models of ROR1-expressing cancers. For switches targeting overlapping or identical epitopes, potency correlated with affinity. Surprisingly, however, we identified a switch targeting a unique epitope with low affinity but mediating potent and selective antitumor activity in vitro and in vivo. Converted to a conventional CAR-T, the same anti-ROR1 mAb (324) outperformed a clinically investigated conventional CAR-T that is based on an anti-ROR1 mAb (R12) with ~200-fold higher affinity. Thus, demonstrating therapeutic utility on their own, sCAR-Ts also facilitate higher throughput screening for the identification of conventional CAR-T candidates for preclinical and clinical studies.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Línea Celular Tumoral , Epítopos , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/metabolismo
8.
Nat Commun ; 12(1): 4077, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34210970

RESUMEN

Emerging data demonstrate that the activity of immune cells can be modulated by microbial molecules. Here, we show that the short-chain fatty acids (SCFAs) pentanoate and butyrate enhance the anti-tumor activity of cytotoxic T lymphocytes (CTLs) and chimeric antigen receptor (CAR) T cells through metabolic and epigenetic reprograming. We show that in vitro treatment of CTLs and CAR T cells with pentanoate and butyrate increases the function of mTOR as a central cellular metabolic sensor, and inhibits class I histone deacetylase activity. This reprogramming results in elevated production of effector molecules such as CD25, IFN-γ and TNF-α, and significantly enhances the anti-tumor activity of antigen-specific CTLs and ROR1-targeting CAR T cells in syngeneic murine melanoma and pancreatic cancer models. Our data shed light onto microbial molecules that may be used for enhancing cellular anti-tumor immunity. Collectively, we identify pentanoate and butyrate as two SCFAs with therapeutic utility in the context of cellular cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Ácidos Grasos Volátiles/metabolismo , Factores Inmunológicos/metabolismo , Inmunoterapia Adoptiva/métodos , Microbiota/fisiología , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Butiratos/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Femenino , Inmunoterapia , Interferón gamma , Subunidad alfa del Receptor de Interleucina-2 , Megasphaera , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos , Receptores Huérfanos Similares al Receptor Tirosina Quinasa , Receptores Acoplados a Proteínas G/genética , Factor de Necrosis Tumoral alfa
9.
Leukemia ; 35(1): 201-214, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32350373

RESUMEN

Multiple myeloma (MM) is incurable, so there is a significant unmet need for effective therapy for patients with relapsed or refractory disease. This situation has not changed despite the recent approval of the anti-CD38 antibody daratumumab, one of the most potent agents in MM treatment. The efficiency of daratumumab might be improved by combining it with synergistic anti-MM agents. We therefore investigated the potential of the histone deacetylase (HDAC) inhibitor ricolinostat to up-regulate CD38 on MM cells, thereby enhancing the performance of CD38-specific therapies. Using quantitative reverse transcription polymerase chain reaction and flow cytometry, we observed that ricolinostat significantly increases CD38 RNA levels and CD38 surface expression on MM cells. Super-resolution microscopy imaging of MM cells by direct stochastic optical reconstruction microscopy confirmed this rise with molecular resolution and revealed homogeneous distribution of CD38 molecules on the cell membrane. Particularly important is that combining ricolinostat with daratumumab induced enhanced lysis of MM cells. We also evaluated next-generation HDAC6 inhibitors (ACY-241, WT-161) and observed similar increase of CD38 levels suggesting that the upregulation of CD38 expression on MM cells by HDAC6 inhibitors is a class effect. This proof-of-concept illustrates the potential benefit of combining HDAC6 inhibitors and CD38-directed immunotherapy for MM treatment.


Asunto(s)
ADP-Ribosil Ciclasa 1/genética , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Glicoproteínas de Membrana/genética , Mieloma Múltiple/genética , ADP-Ribosil Ciclasa 1/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Línea Celular Tumoral , Sinergismo Farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Ácidos Hidroxámicos/farmacología , Inmunofenotipificación , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Pirimidinas/farmacología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
10.
J Immunother Cancer ; 8(2)2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33127653

RESUMEN

BACKGROUND: Immunosuppressive therapy or T-cell depletion in transplant patients can cause uncontrolled growth of Epstein-Barr virus (EBV)-infected B cells resulting in post-transplant lymphoproliferative disease (PTLD). Current treatment options do not distinguish between healthy and malignant B cells and are thereby often limited by severe side effects in the already immunocompromised patients. To specifically target EBV-infected B cells, we developed a novel peptide-selective chimeric antigen receptor (CAR) based on the monoclonal antibody TÜ165 which recognizes an Epstein-Barr nuclear antigen (EBNA)-3C-derived peptide in HLA-B*35 context in a T-cell receptor (TCR)-like manner. In order to attract additional immune cells to proximity of PTLD cells, based on the TÜ165 CAR, we moreover generated T cells redirected for universal cytokine-mediated killing (TRUCKs), which induce interleukin (IL)-12 release on target contact. METHODS: TÜ165-based CAR-T cells (CAR-Ts) and TRUCKs with inducible IL-12 expression in an all-in-one construct were generated. Functionality of the engineered cells was assessed in co-cultures with EBNA-3C-peptide-loaded, HLA-B*35-expressing K562 cells and EBV-infected B cells as PTLD model. IL-12, secreted by TRUCKs on target contact, was further tested for its chemoattractive and activating potential towards monocytes and natural killer (NK) cells. RESULTS: After co-cultivation with EBV target cells, TÜ165 CAR-Ts and TRUCKs showed an increased activation marker expression (CD137, CD25) and release of proinflammatory cytokines (interferon-γ and tumor necrosis factor-α). Moreover, TÜ165 CAR-Ts and TRUCKs released apoptosis-inducing mediators (granzyme B and perforin) and were capable to specifically lyse EBV-positive target cells. Live cell imaging revealed a specific attraction of TÜ165 CAR-Ts around EBNA-3C-peptide-loaded target cells. Of note, TÜ165 TRUCKs with inducible IL-12 showed highly improved effector functions and additionally led to recruitment of monocyte and NK cell lines. CONCLUSIONS: Our results demonstrate that TÜ165 CAR-Ts recognize EBV peptide/HLA complexes in a TCR-like manner and thereby allow for recognizing an intracellular EBV target. TÜ165 TRUCKs equipped with inducible IL-12 expression responded even more effectively and released IL-12 recruited additional immune cells which are generally missing in proximity of lymphoproliferation in immunocompromised PTLD patients. This suggests a new and promising strategy to specifically target EBV-infected cells while sparing and mobilizing healthy immune cells and thereby enable control of EBV-associated lymphoproliferation.


Asunto(s)
Epítopos/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Antígenos HLA-B/metabolismo , Inmunoterapia Adoptiva/métodos , Femenino , Humanos , Masculino
11.
Curr Protoc Immunol ; 128(1): e93, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32150338

RESUMEN

One promising approach to treat hematologic malignancies is the usage of patient-derived CAR T cells. There are continuous efforts to improve the function of these cells, to optimize their receptor, and to use them for the treatment of additional types of cancer and especially solid tumors. In this protocol, an easy and reliable approach for CAR T cell generation is described. T cells are first isolated from peripheral blood (here: leukoreduction system chambers) and afterwards activated for one day with anti-CD3/CD28 Dynabeads. The gene transfer is performed by lentiviral transduction and gene transfer rate can be verified by flowcytometric analysis. Six days after transduction, the stimulatory Dynabeads are removed. T cells are cultured in interleukin-2 conditioned medium for several days for expansion. There is an option to expand CAR T cells further by co-incubation with irradiated, antigen-expressing feeder cell lines. The CAR T cells are ready to use after 10 (without feeder cell expansion) to 24 days (with feeder cell expansion). © 2020 The Authors. Basic Protocol: Generation of CAR T cells by lentiviral transduction.


Asunto(s)
Anticuerpos Monoclonales , Vectores Genéticos , Lentivirus , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transducción Genética , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Biomarcadores , Ingeniería Genética , Vectores Genéticos/genética , Humanos , Separación Inmunomagnética/métodos , Inmunofenotipificación , Inmunoterapia Adoptiva/métodos , Lentivirus/genética , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética
12.
JCI Insight ; 4(18)2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31415244

RESUMEN

Solid tumors impose immunologic and physical barriers to the efficacy of chimeric antigen receptor (CAR) T cell therapy that are not reflected in conventional preclinical testing against singularized tumor cells in 2-dimensional culture. Here, we established microphysiologic three-dimensional (3D) lung and breast cancer models that resemble architectural and phenotypical features of primary tumors and evaluated the antitumor function of receptor tyrosine kinase-like orphan receptor 1-specific (ROR1-specific) CAR T cells. 3D tumors were established from A549 (non-small cell lung cancer) and MDA-MB-231 (triple-negative breast cancer) cell lines on a biological scaffold with intact basement membrane (BM) under static and dynamic culture conditions, which resulted in progressively increasing cell mass and invasive growth phenotype (dynamic > static; MDA-MB-231 > A549). Treatment with ROR1-CAR T cells conferred potent antitumor effects. In dynamic culture, CAR T cells actively entered arterial medium flow and adhered to and infiltrated the tumor mass. ROR1-CAR T cells penetrated deep into tumor tissue and eliminated multiple layers of tumor cells located above and below the BM. The microphysiologic 3D tumor models developed in this study are standardized, scalable test systems that can be used either in conjunction with or in lieu of animal testing to interrogate the antitumor function of CAR T cells and to obtain proof of concept for their safety and efficacy before clinical application.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/terapia , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/inmunología , Receptores Quiméricos de Antígenos/inmunología , Neoplasias de la Mama Triple Negativas/terapia , Alternativas a las Pruebas en Animales , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Anticuerpos de Cadena Única/inmunología , Esferoides Celulares , Linfocitos T/inmunología , Linfocitos T/trasplante , Neoplasias de la Mama Triple Negativas/inmunología
13.
Nat Commun ; 10(1): 3137, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31316055

RESUMEN

Immunotherapy with chimeric antigen receptor-engineered T-cells (CAR-T) is under investigation in multiple myeloma. There are reports of myeloma remission after CD19 CAR-T therapy, although CD19 is hardly detectable on myeloma cells by flow cytometry (FC). We apply single molecule-sensitive direct stochastic optical reconstruction microscopy (dSTORM), and demonstrate CD19 expression on a fraction of myeloma cells (10.3-80%) in 10 out of 14 patients (density: 13-5,000 molecules per cell). In contrast, FC detects CD19 in only 2 of these 10 patients, on a smaller fraction of cells. Treatment with CD19 CAR-T in vitro results in elimination of CD19-positive myeloma cells, including those with <100 CD19 molecules per cell. Similar data are obtained by dSTORM analyses of CD20 expression on myeloma cells and CD20 CAR-T. These data establish a sensitivity threshold for CAR-T and illustrate how super-resolution microscopy can guide patient selection in immunotherapy to exploit ultra-low density antigens.


Asunto(s)
Antígenos CD19/metabolismo , Mieloma Múltiple/terapia , Biomarcadores/metabolismo , Estudios de Cohortes , Citometría de Flujo , Humanos , Inmunoterapia Adoptiva , Interferón gamma/metabolismo , Mieloma Múltiple/metabolismo , Receptores Quiméricos de Antígenos/uso terapéutico
14.
Sci Transl Med ; 11(499)2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31270272

RESUMEN

Immunotherapy with chimeric antigen receptor (CAR)-engineered T cells can be effective against advanced malignancies. CAR T cells are "living drugs" that require technologies to enable physicians (and patients) to maintain control over the infused cell product. Here, we demonstrate that the tyrosine kinase inhibitor dasatinib interferes with the lymphocyte-specific protein tyrosine kinase (LCK) and thereby inhibits phosphorylation of CD3ζ and ζ-chain of T cell receptor-associated protein kinase 70 kDa (ZAP70), ablating signaling in CAR constructs containing either CD28_CD3ζ or 4-1BB_CD3ζ activation modules. As a consequence, dasatinib induces a function-off state in CD8+ and CD4+ CAR T cells that is of immediate onset and can be sustained for several days without affecting T cell viability. We show that treatment with dasatinib halts cytolytic activity, cytokine production, and proliferation of CAR T cells in vitro and in vivo. The dose of dasatinib can be titrated to achieve partial or complete inhibition of CAR T cell function. Upon discontinuation of dasatinib, the inhibitory effect is rapidly and completely reversed, and CAR T cells resume their antitumor function. The favorable pharmacodynamic attributes of dasatinib can be exploited to steer the activity of CAR T cells in "function-on-off-on" sequences in real time. In a mouse model of cytokine release syndrome (CRS), we demonstrated that a short treatment course of dasatinib, administered early after CAR T cell infusion, protects a proportion of mice from otherwise fatal CRS. Our data introduce dasatinib as a broadly applicable pharmacologic on/off switch for CAR T cells.


Asunto(s)
Dasatinib/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Animales , Síndrome de Liberación de Citoquinas/inmunología , Dexametasona/farmacología , Femenino , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Ratones SCID , Fosforilación/efectos de los fármacos
15.
Mol Ther ; 27(2): 287-299, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30573301

RESUMEN

Chimeric antigen receptor (CAR)-T cell immunotherapy is under intense preclinical and clinical investigation, and it involves a rapidly increasing portfolio of novel target antigens and CAR designs. We established a platform that enables rapid and high-throughput CAR-screening campaigns with reporter cells derived from the T cell lymphoma line Jurkat. Reporter cells were equipped with nuclear factor κB (NF-κB) and nuclear factor of activated T cells (NFAT) reporter genes that generate a duplex output of enhanced CFP (ECFP) and EGFP, respectively. As a proof of concept, we modified reporter cells with CD19-specific and ROR1-specific CARs, and we detected high-level reporter signals that allowed distinguishing functional from non-functional CAR constructs. The reporter data were highly reproducible, and the time required for completing each testing campaign was substantially shorter with reporter cells (6 days) compared to primary CAR-T cells (21 days). We challenged the reporter platform to a large-scale screening campaign on a ROR1-CAR library, and we showed that reporter cells retrieved a functional CAR variant that was present with a frequency of only 6 in 1.05 × 106. The data illustrate the potential to implement this reporter platform into the preclinical development path of novel CAR-T cell products and to inform and accelerate the selection of lead CAR candidates for clinical translation.


Asunto(s)
Biblioteca de Genes , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunoterapia/métodos , Células Jurkat , Ratones , Neoplasias/metabolismo , Neoplasias/terapia , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo
17.
Leukemia ; 32(5): 1168-1179, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29472720

RESUMEN

FMS-like tyrosine kinase 3 (FLT3) is a transmembrane protein expressed on normal hematopoietic stem and progenitor cells (HSC) and retained on malignant blasts in acute myeloid leukemia (AML). We engineered CD8+ and CD4+ T-cells expressing a FLT3-specific chimeric antigen receptor (CAR) and demonstrate they confer potent reactivity against AML cell lines and primary AML blasts that express either wild-type FLT3 or FLT3 with internal tandem duplication (FLT3-ITD). We also show that treatment with the FLT3-inhibitor crenolanib leads to increased surface expression of FLT3 specifically on FLT3-ITD+ AML cells and consecutively, enhanced recognition by FLT3-CAR T-cells in vitro and in vivo. As anticipated, we found that FLT3-CAR T-cells recognize normal HSCs in vitro and in vivo, and disrupt normal hematopoiesis in colony-formation assays, suggesting that adoptive therapy with FLT3-CAR T-cells will require subsequent CAR T-cell depletion and allogeneic HSC transplantation to reconstitute the hematopoietic system. Collectively, our data establish FLT3 as a novel CAR target in AML with particular relevance in high-risk FLT3-ITD+ AML. Further, our data provide the first proof-of-concept that CAR T-cell immunotherapy and small molecule inhibition can be used synergistically, as exemplified by our data showing superior antileukemia efficacy of FLT3-CAR T-cells in combination with crenolanib.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/terapia , Receptores Quiméricos de Antígenos/uso terapéutico , Tirosina Quinasa 3 Similar a fms/inmunología , Bencimidazoles/uso terapéutico , Línea Celular Tumoral , Sinergismo Farmacológico , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Piperidinas/uso terapéutico , Receptores Quiméricos de Antígenos/inmunología , Secuencias Repetidas en Tándem/genética , Células Tumorales Cultivadas , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética
18.
J Mol Biol ; 429(19): 2954-2973, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28818634

RESUMEN

Owing to their high affinities and specificities, rabbit monoclonal antibodies (mAbs) have demonstrated value and potential primarily as basic research and diagnostic reagents, but, in some cases, also as therapeutics. To accelerate access to rabbit mAbs bypassing immunization, we generated a large naïve rabbit antibody repertoire represented by a phage display library encompassing >10 billion independent antibodies in chimeric rabbit/human Fab format and validated it by next-generation sequencing. Panels of rabbit mAbs selected from this library against two emerging cancer targets, ROR1 and ROR2, revealed high diversity, affinity, and specificity. Moreover, ROR1- and ROR2-targeting rabbit mAbs demonstrated therapeutic utility as components of chimeric antigen receptor-engineered T cells, further corroborating the value of the naïve rabbit antibody library as a rich and virtually unlimited source of rabbit mAbs.


Asunto(s)
Anticuerpos Monoclonales/aislamiento & purificación , Factores Inmunológicos/aislamiento & purificación , Animales , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Pruebas Inmunológicas de Citotoxicidad , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Factores Inmunológicos/uso terapéutico , Inmunoterapia/métodos , Biblioteca de Péptidos , Conejos , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/uso terapéutico , Linfocitos T/inmunología
19.
Immunobiology ; 222(5): 719-725, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28139257

RESUMEN

Increasing evidence suggests that human NK cells may develop memory-like features. Here, we report the establishment of a robust 8-color flow cytometry panel that allows quantification and functional analysis of different memory-like NK cell subsets (NKG2C+/CD57+, FcεRγ-) from relatively small blood samples. We could confirm previous publications reporting an enhanced prevalence of the mentioned memory-like NK cell subsets in CMV seropositive human donors and were able to show a clear congruence between enhanced expression of NKG2C and CD57, the absence of FcεRγ and CMV seropositivity supporting the hypothesis of memory-like NK cell development following viral infections. While we could not detect significant differences in effector functions (i.e. degranulation and production of IFNγ) in regard to age or CMV seropositivity when looking at the overall NK cell population, a significantly enhanced expression of CD107a and IFNγ could be observed in NKG2C+/CD57+ as well as FcεRγ- NK cell subpopulations in CMV+ donors. This enhancement of effector functions was even more pronounced in NKG2C+/CD57+ NK cells that were also negative for FcεRγ; CMV seropositive donors showed a dramatically increased expression of CD107a as well as IFNγ. With only small-sized volumes of blood needed, our proposed 8-color panel and experimental protocol offers easy handling and a reliable and reproducible option for implementation in accompanying clinical research, e.g. for evaluation of immunosuppressed patients suffering from autoimmune diseases or in allograft recipients.


Asunto(s)
Antígenos CD57/metabolismo , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/inmunología , Citometría de Flujo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Adulto , Anciano , Biomarcadores , Diferenciación Celular , Citocinas/biosíntesis , Femenino , Citometría de Flujo/métodos , Humanos , Memoria Inmunológica , Inmunofenotipificación , Células Asesinas Naturales/citología , Masculino , Persona de Mediana Edad , Receptores de IgE/metabolismo , Adulto Joven
20.
BMC Res Notes ; 7: 710, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25300205

RESUMEN

BACKGROUND: Various kinase inhibitors are known to be ATP-binding cassette (ABC) transporter substrates and resistance acquisition to kinase inhibitors has been associated to increased ABC transporter expression. Here, we investigated the role of the ABC transporters ABCB1, ABCC1, and ABCG2 during melanoma cell resistance acquisition to the V600-mutant BRAF inhibitors PLX4032 (vemurafenib) and PLX4720. PLX4032 had previously been shown to interfere with ABCB1 and ABCG2. PLX4720 had been demonstrated to interact with ABCB1 but to a lower extent than PLX4032. FINDINGS: PLX4032 and PLX4720 affected ABCC1- and ABCG2-mediated drug transport in a similar fashion. In a panel of 16 V600E BRAF-mutated melanoma cell lines consisting of four parental cell lines and their sub-lines with acquired resistance to PLX4032, PLX4720, vincristine (cytotoxic ABCB1 and ABCC1 substrate), or mitoxantrone (cytotoxic ABCG2 substrate), we detected enhanced ABC transporter expression in 4/4 cytotoxic ABC transporter substrate-resistant, 3/4 PLX4720-resistant, and 1/4 PLX4032-resistant melanoma cell lines. CONCLUSION: PLX4032 has the potential to induce ABC transporter expression but this potential is lower than that of PLX4720 or cytotoxic ABC transporter substrates. Since ABC transporters confer multi-drug resistance, this is of relevance for the design of next-line therapies.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/efectos de los fármacos , Antineoplásicos/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Indoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Antineoplásicos/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Indoles/metabolismo , Melanoma/enzimología , Melanoma/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/efectos de los fármacos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Mutación , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Sulfonamidas/metabolismo , Factores de Tiempo , Vemurafenib
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...