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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 29(7): 2239-2252, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-33744469

RESUMEN

MicroRNAs (miRNAs) regulate gene expression by post-transcriptional inhibition of target genes. Proangiogenic small extracellular vesicles (sEVs; popularly identified with the name "exosomes") with a composite cargo of miRNAs are secreted by cultured stem cells and present in human biological fluids. Lipid nanoparticles (LNPs) represent an advanced platform for clinically approved delivery of RNA therapeutics. In this study, we aimed to (1) identify the miRNAs responsible for sEV-induced angiogenesis; (2) develop the prototype of bioinspired "artificial exosomes" (AEs) combining LNPs with a proangiogenic miRNA, and (3) validate the angiogenic potential of the bioinspired AEs. We previously reported that human sEVs from bone marrow (BM)-CD34+ cells and pericardial fluid (PF) are proangiogenic. Here, we have shown that sEVs secreted from saphenous vein pericytes and BM mesenchymal stem cells also promote angiogenesis. Analysis of miRNA datasets available in-house or datamined from GEO identified the let-7 family as common miRNA signature of the proangiogenic sEVs. LNPs with either hsa-let-7b-5p or cyanine 5 (Cy5)-conjugated Caenorhabditis elegans miR-39 (Cy5-cel-miR-39; control miRNA) were prepared using microfluidic micromixing. let-7b-5p-AEs did not cause toxicity and transferred functionally active let-7b-5p to recipient endothelial cells (ECs). let-7b-AEs also improved EC survival under hypoxia and angiogenesis in vitro and in vivo. Bioinspired proangiogenic AEs could be further developed into innovative nanomedicine products targeting ischemic diseases.


Asunto(s)
Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Liposomas/química , MicroARNs/metabolismo , Nanopartículas/química , Neovascularización Fisiológica , Líquido Pericárdico/fisiología , Animales , Exosomas/genética , Vesículas Extracelulares/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Ratones , MicroARNs/genética
2.
Proc Natl Acad Sci U S A ; 113(1): E16-22, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26699502

RESUMEN

Despite progress in systemic small interfering RNA (siRNA) delivery to the liver and to solid tumors, systemic siRNA delivery to leukocytes remains challenging. The ability to silence gene expression in leukocytes has great potential for identifying drug targets and for RNAi-based therapy for leukocyte diseases. However, both normal and malignant leukocytes are among the most difficult targets for siRNA delivery as they are resistant to conventional transfection reagents and are dispersed in the body. We used mantle cell lymphoma (MCL) as a prototypic blood cancer for validating a novel siRNA delivery strategy. MCL is an aggressive B-cell lymphoma that overexpresses cyclin D1 with relatively poor prognosis. Down-regulation of cyclin D1 using RNA interference (RNAi) is a potential therapeutic approach to this malignancy. Here, we designed lipid-based nanoparticles (LNPs) coated with anti-CD38 monoclonal antibodies that are specifically taken up by human MCL cells in the bone marrow of xenografted mice. When loaded with siRNAs against cyclin D1, CD38-targeted LNPs induced gene silencing in MCL cells and prolonged survival of tumor-bearing mice with no observed adverse effects. These results highlight the therapeutic potential of cyclin D1 therapy in MCL and present a novel RNAi delivery system that opens new therapeutic opportunities for treating MCL and other B-cell malignancies.


Asunto(s)
Linfocitos B/inmunología , Linfoma de Células B/terapia , Linfoma de Células del Manto/terapia , Nanomedicina/métodos , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ADP-Ribosil Ciclasa 1/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Ciclina D1/genética , Regulación hacia Abajo , Silenciador del Gen , Humanos , Lípidos , Linfoma de Células B/inmunología , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/inmunología , Ratones , Nanopartículas , ARN Interferente Pequeño/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Ther ; 25(7): 1491-1500, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28392163

RESUMEN

RNAi-based therapy holds great promise, as it can be utilized for the treatment of multiple conditions in an accurate manner via sequence-specific manipulation of gene expression. To date, RNAi therapeutics have advanced into clinical trials for liver diseases and solid tumors; however, delivery of RNAi to leukocytes in general and to lymphocytes in particular remains a challenge. Lymphocytes are notoriously hard to transduce with RNAi payloads and are disseminated throughout the body, often located in deep tissues; therefore, developing an efficient systemic delivery system directed to lymphocytes is not a trivial task. Successful manipulation of lymphocyte function with RNAi possesses immense therapeutic potential, as it will enable researchers to resolve lymphocyte-implicated diseases such as inflammation, autoimmunity, transplant rejection, viral infections, and blood cancers. This potential has propelled the development of novel targeted delivery systems relying on the accumulating research knowledge from multiple disciplines, including materials science and engineering, immunology, and genetics. Here, we will discuss the recent progress in non-viral delivery strategies of RNAi payloads to lymphocytes. Special emphasis will be made on the challenges and potential opportunities in manipulating lymphocyte function with RNAi. These approaches might ultimately become a novel therapeutic modality to treat leukocyte-related diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Linfocitos/efectos de los fármacos , Terapia Molecular Dirigida/métodos , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Animales , Aptámeros de Nucleótidos/administración & dosificación , Aptámeros de Nucleótidos/química , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/terapia , Ensayos Clínicos como Asunto , Dendrímeros/administración & dosificación , Dendrímeros/síntesis química , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Rechazo de Injerto/terapia , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Linfocitos/inmunología , Linfocitos/patología , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Interferente Pequeño/genética , Virosis/genética , Virosis/inmunología , Virosis/patología , Virosis/terapia
4.
Blood ; 123(24): 3797-802, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24778152

RESUMEN

In chronic lymphocytic leukemia (CLL), stimulation of the B-cell receptor (BCR) triggers survival signals. Because in various cells activation of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway provides cells with survival advantage, we wondered whether BCR stimulation activates the JAK/STAT pathway in CLL cells. To stimulate the BCR we incubated CLL cells with anti-IgM antibodies. Anti-IgM antibodies induced transient tyrosine phosphorylation and nuclear localization of phosphorylated (p) STAT3. Immunoprecipitation studies revealed that anti-JAK2 antibodies coimmunoprecipitated pSTAT3 and pJAK2 in IgM-stimulated but not unstimulated CLL cells, suggesting that activation of the BCR induces activation of JAK2, which phosphorylates STAT3. Incubation of CLL cells with the JAK1/2 inhibitor ruxolitinib inhibited IgM-induced STAT3 phosphorylation and induced apoptosis of IgM-stimulated but not unstimulated CLL cells in a dose- and time-dependent manner. Whether ruxolitinib treatment would benefit patients with CLL remains to be determined.


Asunto(s)
Janus Quinasa 2/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Activación de Linfocitos/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Factor de Transcripción STAT3/metabolismo , Anticuerpos Antiidiotipos/farmacología , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Tirosina/metabolismo
5.
J Immunol ; 190(2): 784-93, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23241880

RESUMEN

Emerging data on intraclonal diversity imply that this phenomenon may play a role in the clinical outcome of patients with chronic lymphocytic leukemia (CLL), where subsets of the CLL clone responding more robustly to external stimuli may gain a growth and survival advantage. In this study, we report intraclonal diversity resolved by responses to CD19 engagement in CLL cells, which can be classified into CD19-responsive (CD19-R) and -nonresponive subpopulations. Engagement of CD19 by anti-CD19 Ab rapidly induced cellular aggregation in the CD19-R CLL cells. The CD19-R CLL cells expressed higher surface levels of CD19 and c-myc mRNA, exhibited distinct morphological features, and were preferentially abolished in rituximab-treated patients. Both subpopulations reacted to sIgM stimulation in a similar manner and exhibited similar levels of Akt and Erk phosphorylation, pointing to functional signaling divergence within the BCR. CD19 unresponsiveness was partially reversible, where nonresponding CD19 cells spontaneously recover their signaling capacity following incubation in vitro, pointing to possible in vivo CD19-signaling attenuating mechanisms. This concept was supported by the lower CD19-R occurrence in bone marrow-derived samples compared with cells derived from the peripheral blood of the same patients. CLL patients with >15.25% of the CD19-R cell fraction had a shorter median time to treatment compared with patients with <15.25% of CD19-R cell fraction. In conclusion, divergence in CD19-mediated signaling unfolds both interpatient and intraclonal diversity in CLL. This signaling diversity is associated with physiological implications, including the location of the cells, their responses to anti-CLL therapeutics, and disease progression.


Asunto(s)
Antígenos CD19/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Transducción de Señal , Anticuerpos Monoclonales de Origen Murino/farmacología , Antineoplásicos/farmacología , Células Cultivadas , Colesterol/metabolismo , Progresión de la Enfermedad , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Microdominios de Membrana/química , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Rituximab , Transducción de Señal/efectos de los fármacos
6.
J Immunol ; 188(1): 259-69, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22140262

RESUMEN

Lasting B cell persistence depends on survival signals that are transduced by cell surface receptors. In this study, we describe a novel biological mechanism essential for survival and homeostasis of normal peripheral mature B cells and chronic lymphocytic leukemia cells, regulated by the heparin-binding cytokine, midkine (MK), and its proteoglycan receptor, the receptor-type tyrosine phosphatase ζ (RPTPζ). We demonstrate that MK initiates a signaling cascade leading to B cell survival by binding to RPTPζ. In mice lacking PTPRZ, the proportion and number of the mature B cell population are reduced. Our results emphasize a unique and critical function for MK signaling in the previously described MIF/CD74-induced survival pathway. Stimulation of CD74 with MIF leads to c-Met activation, resulting in elevation of MK expression in both normal mouse splenic B and chronic lymphocytic leukemia cells. Our results indicate that MK and RPTPζ are important regulators of the B cell repertoire. These findings could pave the way toward understanding the mechanisms shaping B cell survival and suggest novel therapeutic strategies based on the blockade of the MK/RPTPζ-dependent survival pathway.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/inmunología , Linfocitos B/inmunología , Citocinas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/inmunología , Receptores de Factores de Crecimiento/inmunología , Transducción de Señal/inmunología , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Midkina , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/inmunología , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/genética , Bazo/inmunología , Bazo/metabolismo
7.
Mol Ther Nucleic Acids ; 35(2): 102224, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38933259

RESUMEN

Locked nucleic acids (LNAs) are a subtype of antisense oligonucleotides (ASOs) that are characterized by a bridge within the sugar moiety. LNAs owe their robustness to this chemical modification, which as the name suggests, locks it in one conformation. This perspective includes two components: a general overview on ASOs from one side and on delivery issues focusing on lipid nanoparticles (LNPs) on the other side. Throughout, a screening of the ongoing clinical trials involving ASOs is given, as well as a take on the versatility and challenges of using LNAs. Finally, we highlight the potential of LNPs as carriers for the successful delivery of LNAs.

8.
Adv Drug Deliv Rev ; 211: 115359, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38857763

RESUMEN

CRISPR/Cas technology presents a promising approach for treating a wide range of diseases, including cancer and genetic disorders. Despite its potential, the translation of CRISPR/Cas into effective in-vivo gene therapy encounters challenges, primarily due to the need for safe and efficient delivery mechanisms. Lipid nanoparticles (LNPs), FDA-approved for RNA delivery, show potential for delivering also CRISPR/Cas, offering the capability to efficiently encapsulate large mRNA molecules with single guide RNAs. However, achieving precise targeting in-vivo remains a significant obstacle, necessitating further research into optimizing LNP formulations. Strategies to enhance specificity, such as modifying LNP structures and incorporating targeting ligands, are explored to improve organ and cell type targeting. Furthermore, the development of base and prime editing technology presents a potential breakthrough, offering precise modifications without generating double-strand breaks (DSBs). Prime editing, particularly when delivered via targeted LNPs, holds promise for treating diverse diseases safely and precisely. This review assesses both the progress made and the persistent challenges faced in using LNP-encapsulated CRISPR-based technologies for therapeutic purposes, with a particular focus on clinical translation.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Lípidos , Nanopartículas , Humanos , Edición Génica/métodos , Nanopartículas/química , Lípidos/química , Animales , Terapia Genética/métodos , Técnicas de Transferencia de Gen , Ácidos Nucleicos/administración & dosificación , Liposomas
9.
Nat Rev Clin Oncol ; 20(11): 739-754, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37587254

RESUMEN

Harnessing mRNA-lipid nanoparticles (LNPs) to treat patients with cancer has been an ongoing research area that started before these versatile nanoparticles were successfully used as COVID-19 vaccines. Currently, efforts are underway to harness this platform for oncology therapeutics, mainly focusing on cancer vaccines targeting multiple neoantigens or direct intratumoural injections of mRNA-LNPs encoding pro-inflammatory cytokines. In this Review, we describe the opportunities of using mRNA-LNPs in oncology applications and discuss the challenges for successfully translating the findings of preclinical studies of these nanoparticles into the clinic. We critically appraise the potential of various mRNA-LNP targeting and delivery strategies, considering physiological, technological and manufacturing challenges. We explore these approaches in the context of the potential clinical applications best suited to each approach and highlight the obstacles that currently need to be addressed to achieve these applications. Finally, we provide insights from preclinical and clinical studies that are leading to this powerful platform being considered the next frontier in oncology treatment.


Asunto(s)
COVID-19 , Nanopartículas , Neoplasias , Humanos , Vacunas contra la COVID-19 , Neoplasias/genética , Neoplasias/terapia , ARN Mensajero/genética , ARN Mensajero/uso terapéutico
10.
Adv Sci (Weinh) ; 10(19): e2301929, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37092557

RESUMEN

Ionizable lipid-based nanoparticles (LNPs) are the most advanced non-viral drug delivery systems for RNA therapeutics and vaccines. However, cell type-specific, extrahepatic mRNA delivery is still a major hurdle, hampering the development of novel therapeutic modalities. Herein, a novel ionizable lipid library is synthesized by modifying hydrophobic tail chains and linkers. Combined with other helper lipids and utilizing a microfluidic mixing approach, stable LNPs are formed. Using Luciferase-mRNA, mCherry mRNA, and Cre mRNA together with a TdTomato animal model, superior lipids forming LNPs for potent cell-type specific mRNA delivery are identified. In vitro assays concluded that combining branched ester tail chains with hydroxylamine linker negatively affects mRNA delivery efficiency. In vivo studies identify Lipid 23 as a liver-trophic, superior mRNA delivery lipid and Lipid 16 as a potent cell type-specific ionizable lipid for the CD11bhi macrophage population without an additional targeting moiety. Finally, in vivo mRNA delivery efficiency and toxicity of these LNPs are compared with SM-102-based LNP (Moderna's LNP formulation) and are shown to be cell-specific compared to SM-102-based LNPs. Overall, this study suggests that a structural combination of tail and linker can drive a novel functionality of LNPs in vivo.


Asunto(s)
Nanopartículas , Animales , ARN Mensajero/genética , Nanopartículas/química , Lípidos/química
11.
Adv Sci (Weinh) ; 10(21): e2301377, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37171801

RESUMEN

Multiple myeloma (MM) is a cancer of differentiated plasma cells that occurs in the bone marrow (BM). Despite the recent advancements in drug development, most patients with MM eventually relapse and the disease remains incurable. RNA therapy delivered via lipid nanoparticles (LNPs) has the potential to be a promising cancer treatment, however, its clinical implementation is limited due to inefficient delivery to non-hepatic tissues. Here, targeted (t)LNPs designed for delivery of RNA payload to MM cells are presented. The tLNPs consist of a novel ionizable lipid and are coated with an anti-CD38 antibody (αCD38-tLNPs). To explore their therapeutic potential, it is demonstrated that LNPs encapsulating small interference RNA (siRNA) against cytoskeleton-associated protein 5 (CKAP5) lead to a ≈90% decrease in cell viability of MM cells in vitro. Next, a new xenograft MM mouse model is employed, which clinically resembles the human disease and demonstrates efficient homing of MM cells to the BM. Specific delivery of αCD38-tLNPs to BM-residing and disseminated MM cells and the improvement in therapeutic outcome of MM-bearing mice treated with αCD38-tLNPs-siRNA-CKAP5 are shown. These results underscore the potential of RNA therapeutics for treatment of MM and the importance of developing effective targeted delivery systems and reliable preclinical models.


Asunto(s)
Mieloma Múltiple , Humanos , Animales , Ratones , Mieloma Múltiple/tratamiento farmacológico , Médula Ósea , Recurrencia Local de Neoplasia , ARN Interferente Pequeño/uso terapéutico
12.
Sci Adv ; 9(14): eade4800, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37018392

RESUMEN

The potential of microtubule-associated protein targets for cancer therapeutics remains largely unexplored due to the lack of target-specific agents. Here, we explored the therapeutic potential of targeting cytoskeleton-associated protein 5 (CKAP5), an important microtubule-associated protein, with CKAP5-targeting siRNAs encapsulated in lipid nanoparticles (LNPs). Our screening of 20 solid cancer cell lines demonstrated selective vulnerability of genetically unstable cancer cell lines in response to CKAP5 silencing. We identified a highly responsive chemo-resistant ovarian cancer cell line, in which CKAP5 silencing led to significant loss in EB1 dynamics during mitosis. Last, we demonstrated the therapeutic potential in an in vivo ovarian cancer model, showing 80% survival rate of siCKAP5 LNPs-treated animals. Together, our results highlight the importance of CKAP5 as a therapeutic target for genetically unstable ovarian cancer and warrants further investigation into its mechanistic aspects.


Asunto(s)
Nanopartículas , Neoplasias Ováricas , Humanos , Animales , Femenino , Silenciador del Gen , Proteínas Asociadas a Microtúbulos/metabolismo , ARN Interferente Pequeño/genética , Microtúbulos/metabolismo , Neoplasias Ováricas/genética
13.
Sci Adv ; 9(10): eadg1036, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36888708

RESUMEN

Messenger RNA (mRNA) lipid nanoparticle (LNP) vaccines have emerged as an effective vaccination strategy. Although currently applied toward viral pathogens, data concerning the platform's effectiveness against bacterial pathogens are limited. Here, we developed an effective mRNA-LNP vaccine against a lethal bacterial pathogen by optimizing mRNA payload guanine and cytosine content and antigen design. We designed a nucleoside-modified mRNA-LNP vaccine based on the bacterial F1 capsule antigen, a major protective component of Yersinia pestis, the etiological agent of plague. Plague is a rapidly deteriorating contagious disease that has killed millions of people during the history of humankind. Now, the disease is treated effectively with antibiotics; however, in the case of a multiple-antibiotic-resistant strain outbreak, alternative countermeasures are required. Our mRNA-LNP vaccine elicited humoral and cellular immunological responses in C57BL/6 mice and conferred rapid, full protection against lethal Y. pestis infection after a single dose. These data open avenues for urgently needed effective antibacterial vaccines.


Asunto(s)
Vacuna contra la Peste , Peste , Yersinia pestis , Ratones , Animales , Peste/prevención & control , Vacuna contra la Peste/genética , Proteínas Bacterianas/genética , Ratones Endogámicos C57BL , Yersinia pestis/genética , Antígenos Bacterianos/genética
14.
Blood ; 115(14): 2852-63, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20154216

RESUMEN

Chronic lymphocytic leukemia (CLL) is the most common leukemia in the Western hemisphere, but its pathogenesis is still poorly understood. Constitutive tyrosine phosphorylation (p) of signal transducer and activator of transcription (STAT) 3 occurs in several solid tumors and hematologic malignancies. In CLL, however, STAT3 is constitutively phosphorylated on serine 727, not tyrosine 705, residues. Because the biologic significance of serine pSTAT3 in CLL is not known, we studied peripheral blood cells of 106 patients with CLL and found that, although tyrosine pSTAT3 was inducible, serine pSTAT3 was constitutive in all patients studied, regardless of blood count, disease stage, or treatment status. In addition, we demonstrated that constitutive serine pSTAT3 translocates to the nucleus by the karyopherin-beta nucleocytoplasmic system and binds DNA. Dephosphorylation of inducible tyrosine pSTAT3 did not affect STAT3-DNA binding, suggesting that constitutive serine pSTAT3 binds DNA. Furthermore, infection of CLL cells with lentiviral STAT3-small hairpin RNA reduced the expression of several STAT3-regulated survival and proliferation genes and induced apoptosis, suggesting that constitutive serine pSTAT3 initiates transcription in CLL cells. Taken together, our data suggest that constitutive phosphorylation of STAT3 on serine 727 residues is a hallmark of CLL and that STAT3 be considered a therapeutic target in this disease.


Asunto(s)
Núcleo Celular/metabolismo , ADN de Neoplasias/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Transcripción STAT3/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular/genética , Anciano , Anciano de 80 o más Años , Apoptosis/genética , Línea Celular Tumoral , Núcleo Celular/genética , Proliferación Celular , Supervivencia Celular/genética , ADN de Neoplasias/genética , Femenino , Humanos , Lentivirus , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/terapia , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Fosforilación/genética , Factor de Transcripción STAT3/genética , Serina/genética , Serina/metabolismo
15.
Ann Hematol ; 91(8): 1271-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22349724

RESUMEN

Chronic lymphocytic leukemia (CLL) cells depend on their microenvironment for proliferation and survival. Ectonucleotidase CD39 has anti-inflammatory properties as it hydrolyzes proinflammatory extracellular ATP, generates anti-inflammatory adenosine, and also protects regulatory T cells from ATP-induced cell death. In this study, we investigated the clinical significance of CD39 expression on CD4(+) T cells in 62 patients with CLL as well as its compartmental regulation and explored the possible mechanisms for its induction. Compared to healthy individuals, CD4(+)CD39(+) lymphocytes were increased in the peripheral blood of patients with CLL and correlated with the advanced stage of disease. CD4(+)CD39(+) cells were also higher in patients with CLL, who needed therapeutic intervention, and in those who had unmutated immunoglobulin heavy chain variable region gene, were ZAP70(+) or had ß2-microglobulin levels of >3 g/L. There were more CD4(+)CD39(+) lymphocytes in the bone marrow compartment than in the peripheral blood, and in vitro studies showed that CD39 can be induced on CD4(+) cells by exposure to ATP or indirectly, following B cell receptor engagement. This may support the notion that the leukemic cells contribute to create an immune-subversive environment, and perhaps to a poorer prognosis. CD39(+) may also serve as a future target for the development of novel therapies with immune-modulating antitumor agents in CLL.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Leucemia Linfocítica Crónica de Células B/diagnóstico , Anciano , Antígenos CD/genética , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Apirasa/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/patología , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Persona de Mediana Edad , Pronóstico , Regulación hacia Arriba/genética
16.
Adv Mater ; 34(13): e2106350, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35044699

RESUMEN

Chemo-immunotherapy is a combination of "standard-of-care" chemotherapy with immunotherapy and it is considered the most advanced therapeutic modality for various types of cancers. However, many cancer patients still poorly respond to current regimen of chemo-immunotherapy and suggest nanotherapeutics as a boosting agent. Recently, heme oxygenase-1 (HO1) is shown to act as an immunotherapeutic molecule in tumor myeloid cells, in addition to general chemoresistance function in cancer cells suggesting that HO1-targeted therapeutics can become a novel, optimal strategy for boosting chemo-immunotherapy in the clinic. Currently the available HO1-inhibitors demonstrate serious adverse effects in clinical use. Herein, tumor myeloid cell- and cancer cell-dual targeted HO1-inhibiting lipid nanotherapeutic boost (T-iLNTB) is developed using RNAi-loaded lipid nanoparticles. T-iLNTB-mediated HO1-inhibition sensitizes cancer cells to "standard-of-care" chemotherapeutics by increasing immunogenic cell death, and directly reprograms tumor myeloid cells with distinguished phenotype. Furthermore, tumor myeloid cell reprogramming by T-iLNTB induces CD8+ cytotoxic T cell recruitment, which drives "Cold-to-Hot" transition and correlates with improved responsiveness to immune checkpoint inhibitor in combination therapy. Finally, ex vivo study proves that HO1-inhibition directly affects tumor macrophage differentiation. This study demonstrates the potential of T-iLNTB as a novel therapeutic modality for boosting chemo-immunotherapy.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Inmunoterapia , Lípidos , Liposomas , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
17.
Invest New Drugs ; 29(5): 1094-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20517635

RESUMEN

BACKGROUND: Constitutive activation of kit contributes to pathogenesis of acute myeloid leukemia (AML) and targeting Kit may be of therapeutic benefit. APcK110, a novel inhibitor of Kit, has potent proapoptotic and antiproliferative activity in AML cell lines and primary AML samples. Here we extend our studies to the activity of APcK110 in a xenograft mouse model. METHODS: After sub-lethal whole body radiation, OCI/AML3 cells were injected intravenously in NOD-SCID mice. Ten days later, either APcK110 or phosphate buffered saline (PBS) was injected intraperitoneally every other day. Kaplan-Meier estimates were used to calculate survival. RESULTS: We show that 1) all mice injected with OCI/AML3 cells developed a clinical and histological picture consistent with myelomonocytic AML; and 2) survival of APcK110-treated mice was significantly longer compared with mice injected with PBS (p = .02). CONCLUSIONS: APcK110 is a novel kit kinase inhibitor with anti-AML activity in vitro and in vivo. Further evaluation in toxicology and clinical studies is warranted.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Proteínas Proto-Oncogénicas c-kit/metabolismo , Análisis de Supervivencia
18.
Invest New Drugs ; 29(6): 1206-12, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20533075

RESUMEN

The prognosis of patients with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) is poor. Chemotherapy is rarely curative and tyrosine kinase inhibitors (TKIs) induce only transient responses. Heat shock protein 90 (Hsp90) is a chaperone protein that is important in signal transduction, cell cycle control, and transcription regulation in both normal and leukemia cells. In the current study, we tested the growth inhibitory and apoptotic effects of a novel Hsp90 inhibitor, EC141 on the Ph+ ALL lines Z-119, Z-181, and Z-33, as well as primary bone marrow-derived blasts from patients with newly diagnosed Ph+ ALL. We found that EC141 inhibited the growth of Ph+ ALL cells in a concentration-dependent manner with IC(50) ranged from 1 to 10 nM. EC141 also inhibited the proliferation of primary bone marrow-derived blasts using the ALL blast colony assay. EC141 down-regulated Hsp90 and up-regulated Hsp70 protein levels, inhibited CrkL phosphorylation, and induced degradation of Bcr-Abl p190 protein through ubiquitin-dependent proteasomal pathway. Furthermore, exposure of Ph+ ALL cells to EC141 resulted in activation of caspase-3, cleavage of poly (ADP-ribose) polymerase (PARP), and induction of apoptosis. In conclusion, our data suggest that EC141 is a potent Hsp90 inhibitor with activity against Ph+ ALL. Further studies to investigate the anticancer effect of EC141 either as a single agent, or in combination in Ph+ ALL and other hematological malignancies are warranted.


Asunto(s)
Proteínas de Fusión bcr-abl/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Proteínas de Fusión bcr-abl/metabolismo , Proteínas HSP70 de Choque Térmico/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Concentración 50 Inhibidora , Poli Adenosina Difosfato Ribosa/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Regulación hacia Arriba/efectos de los fármacos
19.
Adv Mater ; 32(12): e1906128, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31999380

RESUMEN

Lipid nanoparticles (LNPs) are the most advanced nonviral platforms for small interfering RNA (siRNA) delivery that are clinically approved. These LNPs, based on ionizable lipids, are found in the liver and are now gaining much attention in the field of RNA therapeutics. The previous generation of ionizable lipids varies in linker moieties, which greatly influences in vivo gene silencing efficiency. Here novel ionizable amino lipids based on the linker moieties such as hydrazine, hydroxylamine, and ethanolamine are designed and synthesized. These lipids are formulated into LNPs and screened for their efficiency to deliver siRNAs into leukocytes, which are among the hardest to transfect cell types. Two potent lipids based on their in vitro gene silencing efficiencies are also identified. These lipids are further evaluated for their biodistribution profile, efficient gene silencing, liver toxicity, and potential immune activation in mice. A robust gene silencing is also found in primary lymphocytes when one of these lipids is formulated into LNPs with a pan leukocyte selective targeting agent (ß7 integrin). Taken together, these lipids have the potential to open new avenues in delivering RNAs into leukocytes.


Asunto(s)
Lípidos/química , Nanopartículas/química , ARN Interferente Pequeño/metabolismo , Transfección/métodos , Línea Celular Tumoral , Colorantes Fluorescentes/química , Humanos , Cadenas beta de Integrinas/química , Cadenas beta de Integrinas/genética , Cadenas beta de Integrinas/metabolismo , Leucocitos/citología , Leucocitos/metabolismo , Microscopía Fluorescente , Interferencia de ARN , ARN Interferente Pequeño/química
20.
Pharmaceutics ; 12(6)2020 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-32517377

RESUMEN

Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in Western populations. Therapies such as mRNA and siRNA encapsulated in lipid nanoparticles (LNPs) represent a clinically advanced platform and are utilized for a wide variety of applications. Unfortunately, transfection of RNA into CLL cells remains a formidable challenge and a bottleneck for developing targeted therapies for this disease. Therefore, we aimed to elucidate the barriers to efficient transfection of RNA-encapsulated LNPs into primary CLL cells to advance therapies in the future. To this end, we transfected primary CLL patient samples with mRNA and siRNA payloads encapsulated in an FDA-approved LNP formulation and characterized the transfection. Additionally, we tested the potential of repurposing caffeic acid, curcumin and resveratrol to enhance the transfection of nucleic acids into CLL cells. The results demonstrate that the rapid uptake of LNPs is required for successful transfection. Furthermore, we demonstrate that resveratrol enhances the delivery of both mRNA and siRNA encapsulated in LNPs into primary CLL patient samples, overcoming inter-patient heterogeneity. This study points out the important challenges to consider for efficient RNA therapeutics for CLL patients and advocates the use of resveratrol in combination with RNA lipid nanoparticles to enhance delivery into CLL cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA