Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Appl Toxicol ; 44(2): 301-312, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37770382

RESUMEN

Interleukin-12 (IL-12) is known to exert antitumor immune effects by promoting the activation and proliferation of T cells and NK cells within the immune system. However, clinical trials have observed systemic toxicity associated with the administration of IL-12. This has shelved development plans for its use as a cancer therapeutic drug. Therefore, it is critical that we perform a systematic evaluation of the toxicity and safety of repeated IL-12 administration. In this study, we conducted a comprehensive evaluation of the toxicity and safety of repeated rhIL-12 (recombinant human interleukin-12) administration in rhesus monkeys by assessing its effects on the immune system, organ function, and vital signs. Rhesus monkeys were subcutaneously injected with 0.5, 2.5, and 12.5 µg/kg of rhIL-12 for up to for 14 consecutive weeks. The low dose exhibited no signs of toxicity, whereas animals receiving higher doses displayed symptoms such as loose stools, reduced activity, anemia, and elevated liver function indicators (AST and TBIL). Following three administrations of 12.5 µg/kg, high dosing was adjusted to 7.5 µg/kg due to manifestations of symptoms like loose stools, decreased activity, and huddling in the cage. Furthermore, rhesus monkeys exhibited marked immunogenic responses to recombinant human interleukin-12 (rhIL-12). However, based on overall study findings, the No Observed Adverse Effect Level (NOAEL) for the subcutaneous injection of rhIL-12, when repeatedly administered for 3 months in rhesus monkeys, was considered to be 0.5 µg/kg. The Highest Non-Severely Toxic Dose (HNSTD) was considered to be 7.5 µg/kg.


Asunto(s)
Antineoplásicos , Interleucina-12 , Animales , Humanos , Macaca mulatta , Proteínas Recombinantes/toxicidad , Interleucina-12/toxicidad , Células Asesinas Naturales
2.
Exp Hematol Oncol ; 12(1): 85, 2023 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-37777797

RESUMEN

BACKGROUND: Immunosuppression induced by programmed cell death protein 1 (PD1) presents a significant constraint on the effectiveness of chimeric antigen receptor (CAR)-T therapy. The potential of combining PD1/PDL1 (Programmed cell death 1 ligand 1) axis blockade with CAR-T cell therapy is promising. However, developing a highly efficient and minimally toxic approach requires further exploration. Our attempt to devise a novel CAR structure capable of recognizing both tumor antigens and PDL1 encountered challenges since direct targeting of PDL1 resulted in systemic adverse effects. METHODS: In this research, we innovatively engineered novel CARs by grafting the PD1 domain into a conventional second-generation (2G) CAR specifically targeting CD19. These CARs exist in two distinct forms: one with PD1 extramembrane domain (EMD) directly linked to a transmembrane domain (TMD), referred to as PE CAR, and the other with PD1 EMD connected to a TMD via a CD8 hinge domain (HD), known as PE8HT CAR. To evaluate their efficacy, we conducted comprehensive assessments of their cytotoxicity, cytokine release, and potential off-target effects both in vitro and in vivo using tumor models that overexpress CD19/PDL1. RESULTS: The findings of our study indicate that PE CAR demonstrates enhanced cytotoxicity and reduced cytokine release specifically towards CD19 + PDL1 + tumor cells, without off-target effects to CD19-PDL1 + tumor cells, in contrast to 2G CAR-T cells. Additionally, PE CAR showed ameliorative differentiation, exhaustion, and apoptosis phenotypes as assessed by flow cytometry, RNA-sequencing, and metabolic parameter analysis, after encountering CD19 + PDL1 + tumor cells. CONCLUSION: Our results revealed that CAR grafted with PD1 exhibits enhanced antitumor activity with lower cytokine release and no PD1-related off-target toxicity in tumor models that overexpress CD19 and PDL1. These findings suggest that our CAR design holds the potential for effectively addressing the PD1 signal.

3.
Front Pharmacol ; 13: 1011065, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36172182

RESUMEN

Iron oxide nanoparticles (IONPs) are the first generation of nanomaterials approved by the Food and Drug Administration for use as imaging agents and for the treatment of iron deficiency in chronic kidney disease. However, several IONPs-based imaging agents have been withdrawn because of toxic effects and the poor understanding of the underlying mechanisms. This study aimed to evaluate IONPs toxicity and to elucidate the underlying mechanism after intravenous administration in rats. Seven-week-old rats were intravenously administered IONPs at doses of 0, 10, 30, and 90 mg/kg body weight for 14 consecutive days. Toxicity and molecular perturbations were evaluated using traditional toxicological assessment methods and proteomics approaches, respectively. The administration of 90 mg/kg IONPs induced mild toxic effects, including abnormal clinical signs, lower body weight gain, changes in serum biochemical and hematological parameters, and increased organ coefficients in the spleen, liver, heart, and kidneys. Toxicokinetics, tissue distribution, histopathological, and transmission electron microscopy analyses revealed that the spleen was the primary organ for IONPs elimination from the systemic circulation and that the macrophage lysosomes were the main organelles of IONPs accumulation after intravenous administration. We identified 197 upregulated and 75 downregulated proteins in the spleen following IONPs administration by proteomics. Mechanically, the AKT/mTOR/TFEB signaling pathway facilitated autophagy and lysosomal activation in splenic macrophages. This is the first study to elucidate the mechanism of IONPs toxicity by combining proteomics with traditional methods for toxicity assessment.

4.
Front Immunol ; 12: 724211, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34675920

RESUMEN

Chimeric antigen receptor (CAR)-modified T cells targeting CD19 demonstrate unparalleled responses in B cell malignancies. However, high tumor burden limits clinical efficacy and increases the risk of cytokine release syndrome and neurotoxicity, which is associated with over-activation of the CAR-T cells. The hinge domain plays an important role in the function of CAR-T cells. We hypothesized that deletion of glycine, an amino acid with good flexibility, may reduce the flexibility of the hinge region, thereby mitigating CAR-T cell over-activation. This study involved generating a novel CAR by deletion of two consecutive glycine residues in the CD8 hinge domain of second-generation (2nd) CAR, thereafter named 2nd-GG CAR. The 2nd-GG CAR-T cells showed similar efficacy of CAR expression but lower hinge flexibility, and its protein affinity to CD19 protein was lower than that of 2nd CAR-T cells. Compared to the 2nd CAR-T cells, 2nd-GG CAR-T cells reduced proinflammatory cytokine secretion without diminishing the specific cytotoxicity toward tumor cells in vitro. Furthermore, 2nd-GG CAR-T cells prolonged overall survival in an immunodeficient mouse model bearing NALM-6 when tumor burden was high. This study demonstrated that a lower-flexibility of CD8α hinge improved survival under high tumor burden and reduced proinflammatory cytokines in preclinical studies. While there is potential for improved safety and efficacy, yet this needs validation with clinical trials.


Asunto(s)
Antígenos CD8/inmunología , Citocinas/metabolismo , Leucemia Linfocítica Crónica de Células B/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD19/genética , Antígenos CD19/inmunología , Antígenos CD8/genética , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Transfusión de Linfocitos , Ratones , Ratones SCID , Receptores Quiméricos de Antígenos/genética , Análisis de Supervivencia , Linfocitos T/citología , Linfocitos T/trasplante , Transducción Genética , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cytotherapy ; 22(12): 734-743, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32684339

RESUMEN

BACKGROUND AIMS: Chimeric antigen receptor (CAR) T cells have achieved favorable responses in patients with hematologic malignancies, but the outcome has been far from satisfactory in the treatment of tumors with high expression of immunosuppressive molecules. To overcome this limitation, we modified CAR T cells to secrete types of human soluble programmed cell death protein 1 (PD-1) called sPD-1 CAR T cells. METHODS: To compare the effector function between second (conventional second-generation CAR targeting CD19) and sPD-1 CAR T cells, we measured cytotoxicity, cytokine secretion and activation markers incubated with or without tumor cells expressing CD19 and/or programmed cell death ligand 1 (PD-L1). Furthermore, the anti-tumor efficacy of second and sPD-1 CAR T cells was determined using an NSG mouse model bearing NALM-6-PD-L1. Finally, the underlying mechanism was investigated by metabolic parameters and RNA sequencing analysis of different CAR T cells. RESULTS: Compared with second CAR T cells, sPD-1 CAR T cells enhanced killing efficiency toward CD19+PD-L1+ tumor cells in vitro. Furthermore, sPD-1 CAR T cells reduced the tumor burden and prolonged overall survival of the NSG (NOD-SCID-IL2rg) mice bearing NALM-6-PD-L1. To explore the effect of soluble PD-1 on CAR T cells, we found that sPD-1 CAR T cells exhibited higher levels of activation and ameliorative profiles of differentiation, exhaustion, glycolysis and apoptosis. CONCLUSIONS: With constitutive soluble PD-1 secretion, sPD-1 CAR T cells have tended to eradicate tumors with a high expression of PD-L1 more effectively than second CAR T cells. This may be due to soluble PD-1 enhancing apoptosis resistance, aerobic metabolism and a more "stem" differentiation of CAR T cells. Overall, our study presents a feasible strategy to increase the efficacy of CAR T cells.


Asunto(s)
Neoplasias/inmunología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/metabolismo , Animales , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Apoptosis , Antígeno B7-H1/metabolismo , Diferenciación Celular , Femenino , Glucólisis , Humanos , Activación de Linfocitos/inmunología , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Solubilidad
6.
Toxicol Appl Pharmacol ; 284(2): 134-41, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25727309

RESUMEN

OBJECTIVE: To characterize the mechanism of action of thiazolidinedione (TZD)-induced liver mitochondrial toxicity caused by troglitazone, rosiglitazone, and pioglitazone in HepaRG cells. METHODS: Human hepatoma cells (HepaRG) were treated with troglitazone, rosiglitazone, or pioglitazone (12.5, 25, and 50µM) for 48h. The Seahorse Biosciences XF24 Flux Analyzer was used to measure mitochondrial oxygen consumption. The effect of TZDs on reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were detected by flow cytometry. The mitochondrial ultrastructure of HepaRG cells was observed under a transmission electrical microscope (TEM). mtDNA content was evaluated by real-time PCR, and ATP content and mitochondrial respiratory chain (MRC) complex I, II, III, IV activity were measured via chemiluminescence. Results were considered statistically significant at p<0.05. RESULTS: Among the three drugs, troglitazone exhibited the highest potency, followed by rosiglitazone, and then pioglitazone. The TZDs caused varying degrees of mitochondrial respiratory function disorders including decreases in oxygen consumption, MRC activity, and ATP level, and an elevation in ROS level. TZD treatment resulted in mtDNA content decline, reduction in MMP, and alterations of mitochondrial structure. CONCLUSION: All investigated TZDs show a certain degree of mitochondrial toxicity, with troglitazone exhibiting the highest potency. The underlying mechanism of TZD-induced hepatotoxicity may be associated with alterations in mitochondrial respiratory function disorders, oxidative stress, and changes in membrane permeability. These parameters may be used early in drug development to further optimize risk:benefit profiles.


Asunto(s)
Hígado/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Tiazolidinedionas/toxicidad , Línea Celular Tumoral , Cromanos/toxicidad , ADN Mitocondrial/genética , Transporte de Electrón/efectos de los fármacos , Humanos , Hipoglucemiantes/toxicidad , Hígado/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Pioglitazona , Especies Reactivas de Oxígeno/metabolismo , Rosiglitazona , Troglitazona
7.
Proteomics ; 10(16): 2927-41, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20544730

RESUMEN

3Z-3-[(1H-pyrrol-2-yl)-methylidene]-1-(1-piperidinylmethyl)-1,3-2H-indol-2-one (Z24), a synthetic anti-angiogenic compound, inhibits the growth and metastasis of certain tumors. Previous works have shown that Z24 induces hepatotoxicity in rodents. We examined the hepatotoxic mechanism of Z24 at the protein level and looked for potential biomarkers. We used 2-DE and MALDI-TOF/TOF MS to analyze alternatively expressed proteins in rat liver and plasma after Z24 administration. We also examined apoptosis in rat liver and measured levels of intramitochondrial ROS and NAD(P)H redox in liver cells. We found that 22 nonredundant proteins in the liver and 11 in the plasma were differentially expressed. These proteins were involved in several important metabolic pathways, including carbohydrate, lipid, amino acid, and energy metabolism, biotransformation, apoptosis, etc. Apoptosis in rat liver was confirmed with the terminal deoxynucleotidyl transferase dUTP-nick end labeling assay. In mitochondria, Z24 increased the ROS and decreased the NAD(P)H levels. Thus, inhibition of carbohydrate aerobic oxidation, fatty acid beta-oxidation, and oxidative phosphorylation is a potential mechanism of Z24-induced hepatotoxicity, resulting in mitochondrial dysfunction and apoptosis-mediated cell death. In addition, fetub protein and argininosuccinate synthase in plasma may be potential biomarkers of Z24-induced hepatotoxicity.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Mesilatos/toxicidad , Proteoma/efectos de los fármacos , Pirroles/toxicidad , Aminoácidos/metabolismo , Animales , Apoptosis , Proteínas Sanguíneas/análisis , Relación Dosis-Respuesta a Droga , Electroforesis en Gel Bidimensional , Femenino , Histocitoquímica , Etiquetado Corte-Fin in Situ , Hígado/citología , Espectrometría de Masas , NADP/metabolismo , Proteómica/métodos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
8.
Int J Toxicol ; 29(4): 380-90, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20495136

RESUMEN

The current study was designed to investigate the toxicity of 3Z-3-[((1)H-pyrrol-2-yl)-methylidene]-1-(1-piperidinylmethyl)-1, 3-2H-indol-2-one (Z24), a novel synthetic indolin-2-ketone small molecule compound, using toxicogenomic techniques (complementary DNA [cDNA] microarray). Bioinformatic analysis suggested that the main functions of genes with altered expression were consistent with liver cell regeneration, apoptosis, metabolism of energy and fat, and the death receptor (DR)-mediated apoptosis-signaling pathway. Death receptor 4, Bcl-2, Bcl-xl, caspase 3, and cytochrome C, which are involved in the DR-mediated apoptosis-signaling pathway, were altered after Z24 treatment as determined by Western blotting analysis. When hepatocarcinoma cell line (HepG2 cells) treated with Z24 at 0.248 mmol/L for 24 hours, DNA fragmentation reached a maximum, and examination of cell morphology showed typical signs of apoptosis. These results indicate that Z24 can initiate apoptosis in hepatocytes, which in turn causes hepatotoxicity. A possible toxicological mechanism is that apoptosis was induced in hepatocytes by initiating the DR-mediated signal transduction pathway. Apoptosis of hepatocytes might lead to impairment of energy and lipid metabolism and provoke hepatocyte necrosis or inflammation, resulting in hepatotoxicity.


Asunto(s)
Genómica , Hígado/efectos de los fármacos , Mesilatos/toxicidad , Pirroles/toxicidad , Apoptosis/efectos de los fármacos , Secuencia de Bases , Western Blotting , Células Cultivadas , Cartilla de ADN , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Toxicol Appl Pharmacol ; 215(1): 71-82, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16584752

RESUMEN

Antiangiogenic compound has been believed to be an ideal drug in the current cancer biological therapy, but the angiogenesis inhibitors suffer setback for unknown toxicity now. A novel synthetic indolin-s-ketone small molecular compound, 3Z-3-[((1)H-pyrrol-2-yl)-methylidene]-1-(1-piperidinylmethyl)-1,3-2H-indol-2-one (Z24) can inhibit angiogenesis in new blood vessels. The hepatotoxicity effects of Z24 oral administration (dosed at 60, 130 and 200 mg/kg) have been investigated in female Wistar rats by using metabonomic analysis of (1)H NMR spectra of urine, plasma and liver extracts, as well as by clinical chemistry analysis, liver histopathology and electron micrographs examination. The (1)H NMR spectra of the biofluids were analyzed visually and via pattern recognition by using principal component analysis. The metabonomic trajectory analysis on the time-related hepatotoxicity of Z24 was carried out based on the (1)H NMR spectra of urine samples, which were collected daily predose and postdose over an 8-day period. Urinary excretion of citrate, lactate, 2-oxo-glutarate and succinate increased following Z24 dosing. Increased plasma levels of lactate, TMAO and lipid were observed, with concomitant decrease in the level of glucose and phosphatidylcholine. Metabolic profiling on aqueous soluble extracts of liver tissues with the high dose level of Z24 showed an increase in lactate and glutamine, together with a decrease in glucose, glycogen and choline. On the other hand, studies on lipid soluble extracts of liver tissues with the high dose level of Z24 showed increased level in lipid triglycerides and decreased level in unsaturated fatty acids and phosphatidylcholine. Moreover, the most notable effect of Z24 on the metabolism was the reduction in the urinary levels of creatinine and TMAO and the increase in acetate, citrate, succinate and 2-oxo-glutamate with time dependence. The results indicate that in rats Z24 inhibits mitochondrial function through altering the energy and lipid metabolism, which results in the accumulation of free fatty acids and lactate because of the lack of aerobic respiration. These data show that the metabonomic approach represents a promising new technology for the toxicological mechanism study.


Asunto(s)
Mesilatos/toxicidad , Pirroles/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Espectroscopía de Resonancia Magnética , Mesilatos/sangre , Mesilatos/metabolismo , Mesilatos/orina , Microscopía Electrónica de Transmisión , Pirroles/sangre , Pirroles/metabolismo , Pirroles/orina , Ratas
10.
Lin Chuang Er Bi Yan Hou Ke Za Zhi ; 17(11): 652-3, 2003 Nov.
Artículo en Chino | MEDLINE | ID: mdl-14971197

RESUMEN

OBJECTIVE: To explore the characteristic of lacrimal sac fossa on ESS and traditional maxillary operation, and to provide anatomy basis for the endoscopic endonasal dacryocystorhinostomy. METHOD: Twenty-three heads of adult cadaver specimens were examined including the shape, direction and the thickness of wall of lacrimal sac fossa. RESULT: The fossa is formed by the frontal portion of the maxillary bone and the lacrimal bone. The proportion of the frontal portion of the maxillary bone is bigger than the lacrimal bone. There is about 20 degrees between the rear wall of the fossa and the coronal plane. CONCLUSION: Before the endoscopic endonasal dacryocystorhinostomy we should scan the anterior portion of the medial orbital wall so that we can understand the anatomy of the fossa and improve the effect of the surgery.


Asunto(s)
Dacriocistorrinostomía , Aparato Lagrimal/anatomía & histología , Órbita/anatomía & histología , Adulto , Anatomía Regional , Cadáver , Femenino , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA