Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Haematologica ; 108(2): 568-580, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36722406

RESUMO

B-cell maturation antigen (BCMA) is the lead antigen for chimeric antigen receptor (CAR) T-cell therapy in multiple myeloma (MM). A challenge is inter- and intra-patient heterogeneity in BCMA expression on MM cells and BCMA downmodulation under therapeutic pressure. Accordingly, there is a desire to augment and sustain BCMA expression on MM cells in patients that receive BCMA-CAR T-cell therapy. We used all-trans retinoic acid (ATRA) to augment BCMA expression on MM cells and to increase the efficacy of BCMA-CAR T cells in pre-clinical models. We show that ATRA treatment leads to an increase in BCMA transcripts by quantitative reverse transcription polymerase chain reaction and an increase in BCMA protein expression by flow cytometry in MM cell lines and primary MM cells. Analyses with super-resolution microscopy confirmed increased BCMA protein expression and revealed an even distribution of non-clustered BCMA molecules on the MM cell membrane after ATRA treatment. The enhanced BCMA expression on MM cells after ATRA treatment led to enhanced cytolysis, cytokine secretion and proliferation of BCMA-CAR T cells in vitro, and increased efficacy of BCMA-CAR T-cell therapy in a murine xenograft model of MM in vivo (NSG/MM.1S). Combination treatment of MM cells with ATRA and the γ- secretase inhibitor crenigacestat further enhanced BCMA expression and the efficacy of BCMA-CAR T-cell therapy in vitro and in vivo. Taken together, the data show that ATRA treatment leads to enhanced BCMA expression on MM cells and consecutively, enhanced reactivity of BCMA-CAR T cells. The data support the clinical evaluation of ATRA in combination with BCMA-CAR T-cell therapy and potentially, other BCMA-directed immunotherapies.


Assuntos
Secretases da Proteína Precursora do Amiloide , Imunoterapia Adotiva , Mieloma Múltiplo , Tretinoína , Animais , Humanos , Camundongos , Antígeno de Maturação de Linfócitos B , Mieloma Múltiplo/terapia , Linfócitos T , Tretinoína/farmacologia , Receptores de Antígenos Quiméricos
2.
Blood ; 138(19): 1830-1842, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34289026

RESUMO

Acute myeloid leukemia (AML) is an attractive entity for the development of chimeric antigen receptor (CAR) T-cell immunotherapy because AML blasts are susceptible to T-cell-mediated elimination. Here, we introduce sialic acid-binding immunoglobulin-like lectin 6 (Siglec-6) as a novel target for CAR T cells in AML. We designed a Siglec-6-specific CAR with a targeting domain derived from the human monoclonal antibody JML-1. We found that Siglec-6 is commonly expressed on AML cell lines and primary AML blasts, including the subpopulation of AML stem cells. Treatment with Siglec-6 CAR T cells confers specific antileukemia reactivity that correlates with Siglec-6 expression in preclinical models, including induction of complete remission in a xenograft AML model in immunodeficient mice (NSG/U937). In addition, we confirmed Siglec-6 expression on transformed B cells in chronic lymphocytic leukemia (CLL), and specific anti-CLL reactivity of Siglec-6 CAR T cells in vitro. Of particular interest, we found that Siglec-6 is not detectable on normal hematopoietic stem and progenitor cells (HSPCs) and that treatment with Siglec-6 CAR T cells does not affect their viability and lineage differentiation in colony-formation assays. These data suggest that Siglec-6 CAR T-cell therapy may be used to effectively treat AML without the need for subsequent allogeneic hematopoietic stem cell transplantation. In mature normal hematopoietic cells, we detected Siglec-6 in a proportion of memory (and naïve) B cells and basophilic granulocytes, suggesting the potential for limited on-target/off-tumor reactivity. The lack of expression of Siglec-6 on normal HSPCs is a key to differentiating it from other Siglec family members (eg, Siglec-3 [CD33]) and other CAR target antigens (eg, CD123) that are under investigation in AML, and it warrants the clinical investigation of Siglec-6 CAR T-cell therapy.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Imunoterapia Adotiva , Lectinas/imunologia , Leucemia Mieloide Aguda/terapia , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Feminino , Humanos , Imunoterapia Adotiva/métodos , Leucemia Mieloide Aguda/imunologia , Linfócitos T/imunologia , Células U937
3.
Sci Transl Med ; 11(499)2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31270272

RESUMO

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.


Assuntos
Dasatinibe/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/efeitos dos fármacos , Animais , Síndrome da Liberação de Citocina/imunologia , Dexametasona/farmacologia , Feminino , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Camundongos SCID , Fosforilação/efeitos dos fármacos
4.
Adv Cell Gene Ther ; 1(2)2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30420973

RESUMO

OBJECTIVE: Integrins are heterodimeric receptors that convey cell-to-cell and cell-to-matrix interactions. Integrin αvß3 is expressed in several tumour entities including melanoma, glioblastoma, breast, pancreatic and prostate cancer, where it promotes tumour cell survival and metastasis. Here, we generated αvß3-specific chimeric antigen receptor (CAR) T-cells and analysed their antitumour function in pre-clinical models in vitro and in vivo. METHODS: αvß3-CARs comprising a super-humanised hLM609 targeting domain with either high or low affinity (hLM609v7, K d = 3 nM vs. hLM609v11, K d = 160 nM) and equipped with either a long or a short IgG4-Fc extracellular spacer (229 vs. 12 amino acids) were expressed in CD8+ and CD4+ T-cells through lentiviral transduction. RESULTS: αvß3-CAR T-cells eliminated αvß3-positive tumour cells rapidly and specifically, produced IFN-γ and IL-2 (CD4+ > CD8+) and exhibited productive proliferation. In vitro, we observed the strongest reactivity with the higher-affinity hLM609v7 αvß3-CAR in the short spacer configuration, consistent with the tumour membrane-distal localization of the hLM609 epitope. In a murine xenograft model of metastatic A-375 melanoma, the strongest antitumour effect was mediated by the lower-affinity hLM609v11 αvß3-CAR. Notably, a single administration of hLM609v11 αvß3-CAR T-cells was able to induce complete elimination of melanoma lesions, leading to long-term tumour-free survival. CONCLUSIONS: These data establish αvß3 integrin as a novel target for CAR T-cell immunotherapy, and affirm our previous notion that binding domain affinity and spacer length can be calibrated to augment CAR reactivity. CLINICAL IMPLICATIONS: αvß3-CAR T-cells have therapeutic potential in several prevalent solid tumours, including melanoma and triple-negative breast cancer.

5.
Brain Res ; 1616: 58-70, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25960344

RESUMO

The cochlear nucleus is the first brainstem nucleus to receive sensory input from the cochlea. Depriving this nucleus of auditory input leads to cellular and molecular disorganization which may potentially be counteracted by the activation or application of stem cells. Neuronal stem cells (NSCs) have recently been identified in the neonatal cochlear nucleus and a persistent neurogenic niche was demonstrated in this brainstem nucleus until adulthood. The present work investigates whether the neurogenic environment of the cochlear nucleus can promote the survival of engrafted NSCs and whether cochlear nucleus-derived NSCs can differentiate into neurons and glia in brain tissue. Therefore, cochlear nucleus whole-mount explants were co-cultured with NSCs extracted from either the cochlear nucleus or the hippocampus and compared to a second environment using whole-mount explants from the hippocampus. Factors that are known to induce neuronal differentiation were also investigated in these NSC-explant experiments. NSCs derived from the cochlear nucleus engrafted in the brain tissue and differentiated into all cells of the neuronal lineage. Hippocampal NSCs also immigrated in cochlear nucleus explants and differentiated into neurons, astrocytes and oligodendrocytes. Laminin expression was up-regulated in the cochlear nucleus whole-mounts and regulated the in vitro differentiation of NSCs from the cochlear nucleus. These experiments confirm a neurogenic environment in the cochlear nucleus and the capacity of cochlear nucleus-derived NSCs to differentiate into neurons and glia. Consequently, the presented results provide a first step for the possible application of stem cells to repair the disorganization of the cochlear nucleus, which occurs after hearing loss.


Assuntos
Diferenciação Celular/fisiologia , Núcleo Coclear/citologia , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Fator de Crescimento Epidérmico/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Laminina/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
6.
Neuroreport ; 26(6): 309-13, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25714426

RESUMO

The Brn3.1 gene encodes for the protein Brn3.1, which is a member of the POU-IV class of transcription factors. Mutation leads to nonsyndromic human progressive hearing loss (DFNA15). To investigate the suitability of the Brn3.1 promoter for Cre recombinase-induced genetic recombination in cochlear hair cells, we established a transgenic Brn3.1 Cre mouse. This mouse line was crossbred with floxed ROSA26 and ROSA26 reporter mice. The cochleae were histologically analysed in cryosections at E16.5 and whole-mount preparations from P2 until P85. In addition, mice from all used strains and their recombinant offspring were tested electrophysiologically by auditory brainstem responses (ABR) and distorsion product otoacoustic emissions (DPOAE). Cre recombinase activity could be detected in P14 and P21 animals in a mosaic pattern in 26.3 and 9.9% of the outer hair cells, respectively. All investigated mice showed normal ABR and DPOAE values, indicating that neither insertion of the internal ribosome entry site (IRES) Cre cassette into the Brn3.1 gene led to abnormal auditory development nor did the reporter strains show inherited hearing disorders. This study shows that Cre expression under the control of the Brn3.1 promoter is feasible and that the insertion of the internal ribosome entry site Cre cassette into this locus exerted no effects on hearing development. Because of the inconstant pattern and the limited duration of expression, the application of the developed mouse line might be restricted. Also, the unchanged hearing capacity and structural integrity of the organ of Corti in available reporter lines indicate that they may be useful tools for hearing research.


Assuntos
Células Ciliadas Auditivas Externas/fisiologia , Proteínas de Homeodomínio/genética , Integrases/genética , Camundongos Transgênicos/fisiologia , Fator de Transcrição Brn-3C/genética , Animais , Células Ciliadas Auditivas Externas/metabolismo , Camundongos , Camundongos Transgênicos/embriologia , Regiões Promotoras Genéticas
7.
Neuroreport ; 25(12): 960-4, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-24978398

RESUMO

The cochlear nucleus is the first relay station for acoustic information in the auditory pathway and its cellular integrity is affected by hearing loss. Neurotrophic factors, which are known to regulate fundamental processes in the brain, are expressed in the cochlear nucleus and are regulated by the changes in the stimulation. The aim of this study was to evaluate the effect of the neurotrophins Brain derived neurotrophic factor (BDNF) and Neurotrophin 3 (NT-3) and the neurotrophic factor Fibroblast growth factor 2 (FGF2) on primary cultured cells of the mouse cochlear nucleus. No effect on overall cell growth was detected after 8 days in culture by the factors applied. NT-3 had a strong impact on enhancement of neuronal survival, whereas BDNF stimulated neuronal survival and axonal outgrowth. Axonal branching was negatively affected by the administration of BDNF. FGF2 did not show any effect. The results presented represent fundamental research on auditory neurons, but might be one step toward defining novel therapeutic strategies in the future to prevent cochlear nucleus degeneration induced by hearing loss.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Núcleo Coclear/fisiologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento Neural/metabolismo , Neurônios/fisiologia , Animais , Axônios/fisiologia , Crescimento Celular , Sobrevivência Celular/fisiologia , Células Cultivadas , Núcleo Coclear/citologia , Imuno-Histoquímica , Camundongos , Neurônios/citologia
8.
Exp Neurol ; 250: 333-40, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24120439

RESUMO

The progressive motor neuronopathy (pmn/pmn) mouse, an animal model for a fast developing human motor neuron disorder, is additionally characterized by simultaneous progressive sensorineural hearing loss. The gene defect in the pmn/pmn mouse is localized to a missense mutation in the tubulin-specific chaperone E (TBCE) gene on mouse chromosome 13, which is one of the five tubulin-specific chaperons involved in tubulin folding and dimerization. The missense mutation leads to a disturbance of tubulin structures in the auditory nerve and a progressive outer hair cell loss due to apoptosis, which is accompanied by highly elevated ABR-thresholds and loss of DPOAEs. In addition the TBCE protein is selectively expressed in the outer hair cells and the transcellular processes of the inner pillar cells in the cochlea of control and pmn/pmn mouse. We conclude from our study that the mutation of the TBCE gene affects the auditory nerve and the cochlear hair cells simultaneously, leading to progressive hearing loss. This animal model will give the chance to test possible therapeutic strategies in special forms of hearing loss, in which the auditory nerve and the cochlear hair cells are simultaneously affected.


Assuntos
Nervo Coclear/patologia , Células Ciliadas Auditivas Externas/patologia , Perda Auditiva Neurossensorial/patologia , Microtúbulos/patologia , Chaperonas Moleculares/genética , Mutação , Animais , Modelos Animais de Doenças , Eletrofisiologia , Perda Auditiva Neurossensorial/genética , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Mutantes , Doença dos Neurônios Motores/genética , Degeneração Neural/genética , Degeneração Neural/patologia
9.
Exp Brain Res ; 226(3): 393-406, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23455726

RESUMO

Neuronal stem cells have been described in the post-natal cochlear nucleus recently. The aim of the study was to analyse the neurogenic potential in the cochlear nucleus from the early post-natal days until adulthood. Cochlear nuclei from Sprague-Dawley rats from post-natal day P3 up to P40 were examined. Neurosphere assays showed persistent neurosphere formation from the early post-natal days until adulthood. The numbers of generated neurospheres were fewer in older ages. Neurospheres were smaller, but displayed the same pattern of neuronal stem cell markers. The markers GFAP, MBP and ß-III Tubulin showed differentiation of dissociated cells from the neurospheres in all cells of the neuronal lineage. BrdU incorporation could be detected, in an age-dependent decrease, in whole-mount experiments of the cochlear nucleus on all examined days. BrdU co-labelled with Atoh1 and ß-III Tubulin. In addition, gene expression and cellular distribution studies of the neuronal stem cell markers displayed an age-dependent reduction in both quantity and numbers. The presented results display a possible neurogenic potential until adulthood in the cochlear nucleus by in vitro and in vivo experiments. The fact that this potential is highest at a critical period of development reveals possible functional importance for the development of the cochlear nucleus and the auditory function. The persistent neurogenic potential displayed until adulthood could be a neurogenic niche in the adult cochlear nucleus, which might be used for potential therapeutic strategies.


Assuntos
Núcleo Coclear/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Núcleo Coclear/citologia , Núcleo Coclear/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Proteína Básica da Mielina/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...