Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 621(7978): 396-403, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37130545

RESUMEN

Messenger RNA (mRNA) vaccines are being used to combat the spread of COVID-19 (refs. 1-3), but they still exhibit critical limitations caused by mRNA instability and degradation, which are major obstacles for the storage, distribution and efficacy of the vaccine products4. Increasing secondary structure lengthens mRNA half-life, which, together with optimal codons, improves protein expression5. Therefore, a principled mRNA design algorithm must optimize both structural stability and codon usage. However, owing to synonymous codons, the mRNA design space is prohibitively large-for example, there are around 2.4 × 10632 candidate mRNA sequences for the SARS-CoV-2 spike protein. This poses insurmountable computational challenges. Here we provide a simple and unexpected solution using the classical concept of lattice parsing in computational linguistics, where finding the optimal mRNA sequence is analogous to identifying the most likely sentence among similar-sounding alternatives6. Our algorithm LinearDesign finds an optimal mRNA design for the spike protein in just 11 minutes, and can concurrently optimize stability and codon usage. LinearDesign substantially improves mRNA half-life and protein expression, and profoundly increases antibody titre by up to 128 times in mice compared to the codon-optimization benchmark on mRNA vaccines for COVID-19 and varicella-zoster virus. This result reveals the great potential of principled mRNA design and enables the exploration of previously unreachable but highly stable and efficient designs. Our work is a timely tool for vaccines and other mRNA-based medicines encoding therapeutic proteins such as monoclonal antibodies and anti-cancer drugs7,8.


Asunto(s)
Algoritmos , Vacunas contra la COVID-19 , COVID-19 , Estabilidad del ARN , ARN Mensajero , SARS-CoV-2 , Vacunas de ARNm , Animales , Humanos , Ratones , Codón/genética , COVID-19/genética , COVID-19/inmunología , COVID-19/prevención & control , Vacunas contra la COVID-19/química , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/inmunología , Semivida , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/inmunología , Vacunas de ARNm/química , Vacunas de ARNm/genética , Vacunas de ARNm/inmunología , Estabilidad del ARN/genética , Estabilidad del ARN/inmunología , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Mensajero/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/inmunología
2.
PLoS Pathog ; 20(4): e1012116, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38557908

RESUMEN

Since the beginning of the coronavirus disease 2019 (COVID-19) pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, continues to mutate and generates new variants with increasingly severe immune escape, urging the upgrade of COVID-19 vaccines. Here, based on a similar dimeric RBD design as our previous ZF2001 vaccine, we developed a novel broad-spectrum COVID-19 mRNA vaccine, SWIM516, with chimeric Delta-BA.2 RBD dimer delivered by lipopolyplex (LPP). Unlike the popular lipid nanoparticle (LNP), this LPP-delivered mRNA expresses only in the injection site, which avoids potential toxicity to the liver. We demonstrated the broad-spectrum humoral and cellular immunogenicity of this vaccine to Delta and Omicron sub-variants in naïve mice and as booster shots. When challenged with Delta or Omicron live virus, vaccinated human angiotensin-converting enzyme (hACE2) transgenic mice and rhesus macaques were both protected, displaying significantly reduced viral loads and markedly relieved pathological damages. We believe the SWIM516 vaccine qualifies as a candidate for the next-generation broad-spectrum COVID-19 vaccine.


Asunto(s)
COVID-19 , Vacunas de ARNm , Animales , Humanos , Ratones , Vacunas contra la COVID-19 , Macaca mulatta , COVID-19/prevención & control , Inmunización Secundaria , Ratones Transgénicos , ARN Mensajero/genética , SARS-CoV-2/genética , Anticuerpos Neutralizantes , Anticuerpos Antivirales
3.
J Immunol ; 207(12): 3131-3140, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34772699

RESUMEN

The sympathetic nervous system (SNS) is an important regulator of immune cell function during homeostasis and states of inflammation. Recently, the SNS has been found to bolster tumor growth and impair the development of antitumor immunity. However, it is unclear whether the SNS can modulate APC function. Here, we investigated the effects of SNS signaling in murine monocyte-derived macrophages (moMФ) and dendritic cells (DCs) and further combined the nonspecific ß-blocker propranolol with a peptide cancer vaccine for the treatment of melanoma in mice. We report that norepinephrine treatment dramatically altered moMФ cytokine production, whereas DCs were unresponsive to norepinephrine and critically lack ß2-adrenergic receptor expression. In addition, we show that propranolol plus cancer vaccine enhanced peripheral DC maturation, increased the intratumor proportion of effector CD8+ T cells, and decreased the presence of intratumor PD-L1+ myeloid-derived suppressor cells. Furthermore, this combination dramatically reduced tumor growth compared with vaccination alone. Taken together, these results offer insights into the cell-specific manner by which the SNS regulates the APC immune compartment and provide strong support for the use of propranolol in combination with cancer vaccines to improve patient response rates and survival.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Animales , Linfocitos T CD8-positivos , Células Dendríticas , Ratones , Monocitos , Norepinefrina/farmacología , Propranolol/metabolismo , Propranolol/farmacología , Sistema Nervioso Simpático
4.
Semin Immunol ; 34: 114-122, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28947107

RESUMEN

Cancer immunotherapy has become arguably the most promising advancement in cancer research and therapy in recent years. The efficacy of cancer immunotherapy is critically dependent on specific physiological and physical processes - collectively referred to as transport barriers - including the activation of T cells by antigen presenting cells, T cells migration to and penetration into the tumor microenvironment, and movement of nutrients and other immune cells through the tumor microenvironment. Nanotechnology-based approaches have great potential to help overcome these transport barriers. In this review, we discuss the ways that nanotechnology is being leveraged to improve the efficacy and potency of various cancer immunotherapies.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia/métodos , Nanopartículas/uso terapéutico , Nanotecnología , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Presentación de Antígeno , Movimiento Celular , Humanos , Activación de Linfocitos , Neoplasias/inmunología , Microambiente Tumoral
5.
Nanomedicine ; 35: 102400, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33866011

RESUMEN

The production dysregulation of reactive oxygen species (ROS) and nitric oxide (NO) in ischemic tissues results in endothelial dysfunction, hyperinflammation and poor blood circulation. Here, we report a hybrid molecule, SA-10 with both NO donating and ROS scavenging abilities that demonstrated potent cytoprotection and tube formation activity in endothelial cells under H2O2-induced oxidative stress. SA-10 loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (SA-10 NPs) were delivered intramuscularly (IM) to two murine hindlimb ischemia models. In the acute mode ischemia/reperfusion (I/R), the muscle damage, hyperinflammation, and lung edema were significantly reduced 3 days post-dose while in the chronic ischemia model, significant improvement of blood perfusion and physical endurance was observed over 30 days (P < 0.05). Elderly patients with acute and chronic limb ischemia have limited options for surgical or endovascular interventions, so we anticipate that a product like SA-10 NPs has potential as one of the therapeutic alternatives to surgery.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Isquemia/tratamiento farmacológico , Nanocápsulas , Donantes de Óxido Nítrico , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Isquemia/metabolismo , Isquemia/fisiopatología , Ratones , Nanocápsulas/química , Nanocápsulas/uso terapéutico , Donantes de Óxido Nítrico/química , Donantes de Óxido Nítrico/farmacología
6.
Nature ; 508(7494): 103-107, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24670641

RESUMEN

Cancer cells induce a set of adaptive response pathways to survive in the face of stressors due to inadequate vascularization. One such adaptive pathway is the unfolded protein (UPR) or endoplasmic reticulum (ER) stress response mediated in part by the ER-localized transmembrane sensor IRE1 (ref. 2) and its substrate XBP1 (ref. 3). Previous studies report UPR activation in various human tumours, but the role of XBP1 in cancer progression in mammary epithelial cells is largely unknown. Triple-negative breast cancer (TNBC)--a form of breast cancer in which tumour cells do not express the genes for oestrogen receptor, progesterone receptor and HER2 (also called ERBB2 or NEU)--is a highly aggressive malignancy with limited treatment options. Here we report that XBP1 is activated in TNBC and has a pivotal role in the tumorigenicity and progression of this human breast cancer subtype. In breast cancer cell line models, depletion of XBP1 inhibited tumour growth and tumour relapse and reduced the CD44(high)CD24(low) population. Hypoxia-inducing factor 1α (HIF1α) is known to be hyperactivated in TNBCs. Genome-wide mapping of the XBP1 transcriptional regulatory network revealed that XBP1 drives TNBC tumorigenicity by assembling a transcriptional complex with HIF1α that regulates the expression of HIF1α targets via the recruitment of RNA polymerase II. Analysis of independent cohorts of patients with TNBC revealed a specific XBP1 gene expression signature that was highly correlated with HIF1α and hypoxia-driven signatures and that strongly associated with poor prognosis. Our findings reveal a key function for the XBP1 branch of the UPR in TNBC and indicate that targeting this pathway may offer alternative treatment strategies for this aggressive subtype of breast cancer.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Animales , Antígeno CD24/metabolismo , Hipoxia de la Célula/genética , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Silenciador del Gen , Humanos , Receptores de Hialuranos/metabolismo , Ratones , Invasividad Neoplásica , Recurrencia Local de Neoplasia , Pronóstico , ARN Polimerasa II/metabolismo , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transcripción Genética , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Neoplasias de la Mama Triple Negativas/genética , Respuesta de Proteína Desplegada , Proteína 1 de Unión a la X-Box
7.
Biomed Microdevices ; 21(2): 39, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30949852

RESUMEN

Effective migration of dendritic cells into the lymphatic system organs is the prerequisite for a functional dendritic cell vaccine. We have previously developed a porous silicon microparticle (PSM)-based therapeutic dendritic cell vaccine (Nano-DC vaccine) where PSM serves both as the vehicle for antigen peptides and an adjuvant. Here, we analyzed parameters that determined dendritic cell uptake of PSM particles and Nano-DC vaccine accumulation in lymphatic tissues in a murine model of HER2-positive breast cancer. Our study revealed a positive correlation between sphericity of the PSM particles and their cellular uptake by circulating dendritic cells. In addition, the intravenously administered vaccines accumulated more in the spleens and inguinal lymph nodes, while the intradermally inoculated vaccines got enriched in the popliteal lymph nodes. Furthermore, mice with large tumors received more vaccines in the lymph nodes than those with small to medium size tumors. Information from this study will provide guidance on design and optimization of future therapeutic cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/metabolismo , Células Dendríticas/metabolismo , Nanomedicina , Animales , Transporte Biológico , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Células Dendríticas/inmunología , Ratones , Microesferas , Fagocitos/inmunología , Silicio/química , Distribución Tisular , Carga Tumoral/inmunología
8.
Chem Soc Rev ; 47(6): 1969-1995, 2018 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-29417968

RESUMEN

Cancer treatment still faces a lot of obstacles such as tumor heterogeneity, drug resistance and systemic toxicities. Beyond the traditional treatment modalities, exploitation of RNA interference (RNAi) as an emerging approach has immense potential for the treatment of various gene-caused diseases including cancer. The last decade has witnessed enormous research and achievements focused on RNAi biotechnology. However, delivery of small interference RNA (siRNA) remains a key challenge in the development of clinical RNAi therapeutics. Indeed, functional nanomaterials play an important role in siRNA delivery, which could overcome a wide range of sequential physiological and biological obstacles. Nanomaterial-formulated siRNA systems have potential applications in protection of siRNA from degradation, improving the accumulation in the target tissues, enhancing the siRNA therapy and reducing the side effects. In this review, we explore and summarize the role of functional inorganic-organic hybrid systems involved in the siRNA therapeutic advancements. Additionally, we gather the surface engineering strategies of hybrid systems to optimize for siRNA delivery. Major progress in the field of inorganic-organic hybrid platforms including metallic/non-metallic cores modified with organic shells or further fabrication as the vectors for siRNA delivery is discussed to give credit to the interdisciplinary cooperation between chemistry, pharmacy, biology and medicine.


Asunto(s)
Sistemas de Liberación de Medicamentos , ARN Interferente Pequeño/uso terapéutico , Animales , Humanos , Tamaño de la Partícula , ARN Interferente Pequeño/química
9.
Mol Pharm ; 15(5): 1814-1825, 2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29537266

RESUMEN

Selective drug accumulation in the malignant tissue is a prerequisite for effective cancer treatment. However, most drug molecules and their formulated particles are blocked en route to the destiny tissue due to the existence of multiple biological and physical barriers including the tumor microvessel endothelium. Since the endothelial cells on the surface of the microvessel wall can be modulated by inflammatory cytokines and chemokines secreted by the tumor or stromal cells, an effective drug delivery approach is to enhance interaction between the drug particles and the unique spectrum of surface proteins on the tumor endothelium. In this study, we performed in vivo screening for thioaptamers that bind to the bone marrow endothelium with specificity in a murine model of lymphoma with bone marrow involvement (BMI). The R1 thioaptamer was isolated based on its high homing potency to bones with BMI, and 40-60% less efficiency in accumulation to healthy bones. In cell culture, R1 binds to human umbilical vein endothelial cells (HUVEC) with a high affinity ( Kd ≈ 3 nM), and the binding affinity can be further enhanced when cells were treated with a mixture of lymphoma cell and bone marrow cell conditioned media. Cellular uptake of R1 is through clathrin-mediated endocytosis. Conjugating R1 on to the surface of liposomal doxorubicin nanoparticles resulted in 2-3-fold increase in drug accumulation in lymphoma BMI. Taking together, we have successfully identified a thioaptamer that preferentially binds to the endothelium of lymphoma BMI. It can serve as an affinity moiety for targeted delivery of drug particles to the disease organ.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Células de la Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de los fármacos , ADN/administración & dosificación , Linfoma/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Animales , Línea Celular , Línea Celular Tumoral , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones SCID , Polietilenglicoles/farmacología
10.
Nano Lett ; 17(5): 2913-2920, 2017 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-28418672

RESUMEN

Ruthenium coordination complexes have the potential to serve as novel theranostic agents for cancer. However, a major limitation in their clinical implementation is effective tumor accumulation. In this study, we have developed a liposome-based theranostic nanodelivery system for [Ru(phen)2dppz](ClO4)2 (Lipo-Ru). This ruthenium polypyridine complex emits a strong fluorescent signal when incorporated in the hydrophobic lipid bilayer of the delivery vehicle or in the DNA helix, enabling visualization of the therapeutic agent in tumor tissues. Incubation of MDA-MB-231 breast cancer cells with Lipo-Ru induced double-strand DNA breaks and triggers apoptosis. In a mouse model of triple-negative breast cancer, treatment with Lipo-Ru dramatically reduced tumor growth. Biodistribution studies of Lipo-Ru revealed that more than 20% of the injected dose accumulated in the tumor. These results suggest that Lipo-Ru could serve as a promising theranostic platform for cancer.


Asunto(s)
Antineoplásicos/química , Complejos de Coordinación/química , Portadores de Fármacos/química , Compuestos Heterocíclicos con 3 Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/química , Piridinas/química , Rutenio/química , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/administración & dosificación , Femenino , Humanos , Liposomas , Ratones , Ratones Desnudos , Nanomedicina Teranóstica , Distribución Tisular
11.
Int J Mol Sci ; 19(4)2018 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-29596317

RESUMEN

Feline injection site sarcomas (FISS) are malignant skin tumors with high recurrence rates despite the primary treatment of radical surgical resections. Adjunctive radiotherapy or chemotherapy with doxorubicin is mostly ineffective. Cellular and molecular causes of multidrug resistance, specific physio-chemical properties of solid tumors impairing drug transport, and the tumor microenvironment have been indicated for causing standard chemotherapy failure. Gold nanoparticles are promising imaging tools, nanotherapeutics, and drug delivery systems (DDS) for chemotherapeutics, improving drug transport within solid tumors. This study was conducted to assess the distribution of 4-nm glutathione-stabilized gold nanoparticles in FISS and their influence on kidney and liver parameters in nude mice. The role of gold nanoparticles as a doxorubicin DDS in FISS was examined to determine the potential reasons for failure to translate results from in vitro to in vivo studies. Grade III tumors characterized by a large area of necrosis at their core displayed positive immuneexpression of tumor-associated macrophages (TAM) at both the periphery and within the tumor core near the area of necrosis. Gold nanoparticles did not cause necrosis at the injection site and had no negative effect on liver and kidney parameters in nude mice. Gold nanoparticles accumulated in the tumor core and at the periphery and co-internalized with TAM-an important observation and potential therapeutic target warranting further investigation. The large area of necrosis and high immunoexpression of TAM, indicating "pro-tumor macrophages", may be responsible for FISS tumor progression and therapeutic failure. However, further studies are required to test this hypothesis.


Asunto(s)
Antibióticos Antineoplásicos , Doxorrubicina , Sistemas de Liberación de Medicamentos/métodos , Glutatión , Oro , Nanopartículas del Metal , Sarcoma , Neoplasias Cutáneas , Animales , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Glutatión/química , Glutatión/farmacocinética , Glutatión/farmacología , Oro/química , Oro/farmacología , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Ratones , Ratones Desnudos , Sarcoma/tratamiento farmacológico , Sarcoma/metabolismo , Sarcoma/patología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Blood ; 125(19): 2968-73, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25712988

RESUMEN

Tyrosine kinase inhibitors (TKIs) are used as a frontline therapy for BCR-ABL(+) acute lymphoblastic leukemia (ALL). However, resistance to TKI therapy arises rapidly, and its underlying molecular mechanisms are poorly understood. In this study, we identified a novel cascade of events initiated by TKIs and traversing through mesenchymal stem cells (MSCs) to leukemic cells, leading to resistance. MSCs exposed to TKIs acquired a new functional status with the expression of genes encoding for chemo-attractants, adhesion molecules, and prosurvival growth factors, and this priming enabled leukemic cells to form clusters underneath the MSCs. This cluster formation was associated with the protection of ALL cells from therapy as leukemic cells switched from BCR-ABL signaling to IL-7R/Janus kinase signaling to survive in the MSC milieu. Our findings illustrate a novel perspective in the evolution of TKI resistance and provide insights for advancing the treatment of BCR-ABL(+) ALL.


Asunto(s)
Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/metabolismo , Células Madre Mesenquimatosas/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Proteínas de Fusión bcr-abl/genética , Perfilación de la Expresión Génica , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
13.
PLoS Comput Biol ; 12(6): e1004969, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27286441

RESUMEN

AUTHOR SUMMARY: Cancer treatment efficacy can be significantly enhanced through the elution of drug from nano-carriers that can temporarily stay in the tumor vasculature. Here we present a relatively simple yet powerful mathematical model that accounts for both spatial and temporal heterogeneities of drug dosing to help explain, examine, and prove this concept. We find that the delivery of systemic chemotherapy through a certain form of nano-carriers would have enhanced tumor kill by a factor of 2 to 4 over the standard therapy that the patients actually received. We also find that targeting blood volume fraction (a parameter of the model) through vascular normalization can achieve more effective drug delivery and tumor kill. More importantly, this model only requires a limited number of parameters which can all be readily assessed from standard clinical diagnostic measurements (e.g., histopathology and CT). This addresses an important challenge in current translational research and justifies further development of the model towards clinical translation.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Animales , Biología Computacional , Simulación por Computador , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/uso terapéutico , Femenino , Ratones , Ratones Endogámicos BALB C , Nanopartículas/uso terapéutico , Análisis Espacio-Temporal
14.
Proc Natl Acad Sci U S A ; 111(24): 8838-43, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24876273

RESUMEN

We previously described a gene signature for breast cancer stem cells (BCSCs) derived from patient biopsies. Selective shRNA knockdown identified ribosomal protein L39 (RPL39) and myeloid leukemia factor 2 (MLF2) as the top candidates that affect BCSC self-renewal. Knockdown of RPL39 and MLF2 by specific siRNA nanoparticles in patient-derived and human cancer xenografts reduced tumor volume and lung metastases with a concomitant decrease in BCSCs. RNA deep sequencing identified damaging mutations in both genes. These mutations were confirmed in patient lung metastases (n = 53) and were statistically associated with shorter median time to pulmonary metastasis. Both genes affect the nitric oxide synthase pathway and are altered by hypoxia. These findings support that extensive tumor heterogeneity exists within primary cancers; distinct subpopulations associated with stem-like properties have increased metastatic potential.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Pulmonares/genética , Células Madre Neoplásicas/citología , Óxido Nítrico Sintasa/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Ribosómicas/metabolismo , Animales , Neoplasias de la Mama/prevención & control , Línea Celular Tumoral , Movimiento Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hipoxia , Neoplasias Pulmonares/metabolismo , Ratones , Ratones SCID , Mutación , Metástasis de la Neoplasia , Trasplante de Neoplasias , Óxido Nítrico/química , Óxido Nítrico Sintasa/antagonistas & inhibidores , ARN Interferente Pequeño/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Factores de Tiempo
15.
Pharmacol Res ; 113(Pt A): 92-99, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27546164

RESUMEN

Various nanodelivery systems have been designed to release therapeutic agents upon contact with specific enzymes. However, enzyme-triggered release typically takes place in the tissue interstitium, thereby resulting in the extracellular delivery of drugs. Here, we have designed an enzyme-stimulated multistage vector (ESMSV), which enables stimulus-triggered release of drug-encapsulated nanoparticles from a microparticle. Specifically, polymeric nanoparticles with a surface matrix metalloproteinase-2 (MMP2) peptide substrate were conjugated to the surface of porous silicon microparticles. In the presence of MMP2, the polymeric nanoparticles were released into the tumor interstitium. This platform can be used to attain triggered drug release, while simultaneously facilitating the cellular internalization of drugs. The results indicate that nanoparticle release was MMP2-specific and resulted in improved intracellular uptake of hydrophobic agents in the presence of MMP2. Furthermore, in a mouse model of melanoma lung metastasis, systemic delivery of ESMSVs caused a substantial increase in intracellular accumulation of agents in cancer cells in comparison to delivery with non-stimulus-responsive particles.


Asunto(s)
Portadores de Fármacos/química , Enzimas/química , Neoplasias/tratamiento farmacológico , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/química , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Nanopartículas/química , Tamaño de la Partícula , Polietilenglicoles/química , Polímeros/química
16.
Pharmacol Res ; 111: 413-421, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27394167

RESUMEN

Cancer cells have high rates of glycolysis and lactic acid fermentation in order to fuel accelerated rates of cell division (Warburg effect). Here, we present a strategy for merging cancer and yeast metabolism to remove pyruvate, a key intermediate of cancer cell metabolism, and produce the toxic compound acetaldehyde. This approach was achieved by administering the yeast enzyme pyruvate decarboxylase to triple negative breast cancer cells. To overcome the challenges of protein delivery, a nanoparticle-based system consisting of cationic lipids and porous silicon were employed to obtain efficient intracellular uptake. The results demonstrate that the enzyme therapy decreases cancer cell viability through production of acetaldehyde and reduction of lactic acid fermentation.


Asunto(s)
Antineoplásicos/farmacología , Metabolismo Energético/efectos de los fármacos , Piruvato Descarboxilasa/farmacología , Proteínas de Saccharomyces cerevisiae/farmacología , Saccharomyces cerevisiae/enzimología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Acetaldehído/metabolismo , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos , Composición de Medicamentos , Femenino , Fermentación , Glucólisis , Humanos , Ácido Láctico/metabolismo , Lípidos/química , Nanopartículas , Porosidad , Piruvato Descarboxilasa/química , Piruvato Descarboxilasa/aislamiento & purificación , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Silicio/química , Neoplasias de la Mama Triple Negativas/enzimología , Neoplasias de la Mama Triple Negativas/patología
17.
Small ; 10(7): 1250-4, 2014 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24323884

RESUMEN

A novel function of polyarginine as an activator of the immune system is reported. Single-molecule fluorescence imaging and single-molecule force measurements demonstrate that polyarginine binds specifically to the toll-like receptor 4 (TLR4), inducing receptor dimerization and activation. Moreover, polyarginine induced immune activation inhibits tumor growth. These results suggest the potential use of polyarginine for cancer immunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Inmunidad/efectos de los fármacos , Neoplasias/inmunología , Péptidos/farmacología , Receptor Toll-Like 4/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fluorescencia , Interferones/genética , Interferones/metabolismo , Ratones Endogámicos C57BL , Neoplasias/patología , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Termodinámica , Receptor Toll-Like 4/deficiencia
18.
Clin Chem ; 60(1): 233-42, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24146311

RESUMEN

BACKGROUND: Carboxypeptidase N (CPN) is important in regulating vasoactive peptide hormones, growth factors, and cytokines by specifically cleaving their C-terminal basic residues. We investigated whether circulating peptides specifically cleaved by CPN in the tumor microenvironment can be stage-specific indicators of breast cancer. METHODS: CPN activity was measured using an ex vivo peptide cleavage assay by incubating synthesized C3f peptide (His6-C3f_S1304-R1320-His6) in interstitial fluids of breast tumors and adjacent normal breast tissues in mice with orthotopic implantation of the human cell line MDA-MB-231. The nature and extent of peptide cleavage by CPN was investigated by fragment profiling using nanopore fractionation and mass spectrometry. The fragment profiles in interstitial fluid correlated with concentrations of CPN-catalyzed peptides in blood samples taken from the tumor-bearing mice, healthy women, and breast cancer patients. CPN expression in the same set of samples was further examined by immunohistochemistry and immunoblotting. RESULTS: We showed that generation of C3f_R1310-L1319 specifically correlated with the CPN expression level. In both the mouse and clinical patient samples, CPN was clearly increased in tumor tissues compared with normal breast tissue, whereas corresponding CPN abundance in blood remained constant. Concentrations of 6 CPN-catalyzed peptides predominantly increased in sera taken from the mice (n = 8) at 2 weeks after orthotopic implantation. Six homologous peptides displayed significantly higher expression in the patients' plasma as early as the first pathologic stage of breast cancer. CONCLUSIONS: Circulating CPN-catalyzed peptide concentrations reflect the CPN activity in tumors. These biomarkers show strong potential for the noninvasive and early diagnosis of breast cancer.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias de la Mama/diagnóstico , Detección Precoz del Cáncer , Lisina Carboxipeptidasa/sangre , Péptidos/sangre , Animales , Biomarcadores de Tumor/antagonistas & inhibidores , Neoplasias de la Mama/enzimología , Modelos Animales de Enfermedad , Femenino , Humanos , Lisina Carboxipeptidasa/metabolismo , Ratones , Péptidos/antagonistas & inhibidores , Peptidomiméticos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Microambiente Tumoral
19.
Phys Biol ; 11(6): 065002, 2014 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-25427073

RESUMEN

There is substantial heterogeneity in the clinical behavior of pancreatic cancer and in its response to therapy. Some of this variation may be due to differences in delivery of cytotoxic therapies between patients and within individual tumors. Indeed, in 12 patients with resectable pancreatic cancer, we previously demonstrated wide inter-patient variability in the delivery of gemcitabine as well as in the mass transport properties of tumors as measured by computed tomography (CT) scans. However, the variability of drug delivery and transport properties within pancreatic tumors is currently unknown. Here, we analyzed regional measurements of gemcitabine DNA incorporation in the tumors of the same 12 patients to understand the degree of intra-tumoral heterogeneity of drug delivery. We also developed a volumetric segmentation approach to measure mass transport properties from the CT scans of these patients and tested inter-observer agreement with this new methodology. Our results demonstrate significant heterogeneity of gemcitabine delivery within individual pancreatic tumors and across the patient cohort, with gemcitabine DNA incorporation in the inner portion of the tumors ranging from 38 to 74% of the total. Similarly, the CT-derived mass transport properties of the tumors had a high degree of heterogeneity, ranging from minimal difference to almost 200% difference between inner and outer portions of the tumor. Our quantitative method to derive transport properties from CT scans demonstrated less than 5% difference in gemcitabine prediction at the average CT-derived transport value across observers. These data illustrate significant inter-patient and intra-tumoral heterogeneity in the delivery of gemcitabine, and highlight how this variability can be reproducibly accounted for using principles of mass transport. With further validation as a biophysical marker, transport properties of tumors may be useful in patient selection for therapy and prediction of therapeutic outcome.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Desoxicitidina/análogos & derivados , Sistemas de Liberación de Medicamentos , Neoplasias Pancreáticas/metabolismo , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/metabolismo , Transporte Biológico , Aductos de ADN/metabolismo , ADN de Neoplasias/metabolismo , Desoxicitidina/administración & dosificación , Desoxicitidina/metabolismo , Desoxicitidina/farmacocinética , Humanos , Inyecciones Intravenosas , Pancreatectomía , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Distribución Tisular , Tomógrafos Computarizados por Rayos X , Microambiente Tumoral , Gemcitabina
20.
Haematologica ; 99(10): 1591-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24997151

RESUMEN

The transcription factor Sox4 plays an indispensable role in the development of early progenitor B cells from hematopoietic stem cells. However, its role in B-cell acute lymphoblastic leukemia, a malignant counterpart of normal progenitor B cells, is not fully understood. Here we show that SOX4 is highly expressed in human acute lymphoblastic leukemia cells. To systematically study the function of Sox4 in acute lymphoblastic leukemia, we established a genetically defined mouse leukemia model by transforming progenitor B cells carrying a floxed Sox4 allele and inducing deletion of the allele by the self-excising Cre recombinase. This model allowed us to work with two groups of leukemic cells that had either one copy or both copies of Sox4 deleted. We found that depletion of Sox4 in transformed cells in vitro reduced cell growth in vitro and the progression of leukemia in vivo. Moreover, depletion of Sox4 in leukemic cells in vivo prolonged the survival of the mice, suggesting that it could be a potential target in acute lymphoblastic leukemia therapy. Our microarray and bioChIP studies revealed that Tcf7l1 was the key gene directly regulated by Sox4. Knockdown of Tcf7l1 reduced cell proliferation, just as did knockout of Sox4, and ectopic expression of Tcf7l1 could reverse the effect of Sox4 knockout on cell proliferation. These data suggest that Sox4 and Tcf7l1 form a functional axis that promotes the progression of BCR-ABL-positive acute lymphoblastic leukemia.


Asunto(s)
Proteínas de Fusión bcr-abl/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Factores de Transcripción SOXC/metabolismo , Proteína 1 Similar al Factor de Transcripción 7/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Análisis por Conglomerados , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción SOXC/genética , Proteína 1 Similar al Factor de Transcripción 7/genética , Carga Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA