Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Bioconjug Chem ; 35(2): 187-202, 2024 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-38318778

RESUMEN

To meet the current need for a tumor-selective, targeted therapy regimen associated with reduced toxicity, our laboratory has developed a spontaneously assembled nanostructure that resembles high-density lipoproteins (HDLs). These myristoyl-5A (MYR-5A) nanotransporters are designed to safely transport lipophilic pharmaceuticals, including a novel anthracycline drug (N-benzyladriamycin-14-valerate (AD198)). This formulation has been found to enhance the therapeutic efficacy and reduced toxicity of drugs in preclinical studies of 2D and 3D models of Ewing sarcoma (EWS) and cardiomyocytes. Our findings indicate that the MYR-5A/AD198 nanocomplex delivers its payload selectively to cancer cells via the scavenger receptor type B1 (SR-B1), thus providing a solid proof of concept for the development of an improved and highly effective, potentially personalized therapy for EWS while protecting against treatment-associated cardiotoxicity.


Asunto(s)
Doxorrubicina/análogos & derivados , Sarcoma de Ewing , Humanos , Sarcoma de Ewing/tratamiento farmacológico , Nanoconjugados/uso terapéutico , Antraciclinas/farmacología , Antraciclinas/uso terapéutico , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular Tumoral
2.
Appl Biochem Biotechnol ; 196(1): 145-159, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37103734

RESUMEN

Chronic myeloid leukemia (CML) as a bone marrow stem cell clonal disease appears from the proliferation of granulocyte cells at all stages of maturation. If the disease diagnosis is not early, patients enter the blastic phase, which decreases their survival rate to 3-6 months. It implies the significance of the early diagnosis of CML. In this study, we introduce a simple array for diagnosis of the K562 cells as the human immortalized myeloid leukemia cell line. The developed aptamer-based biosensor (aptasensor) includes the T2-KK1B10 aptamer strands attached to the surface of mesoporous silica nanoparticles (MSNPs) with the cavities accumulated from rhodamine B and coated by both Ca2+ ions and ATP aptamer. The aptamer-based nanoconjugate can enter the K562 cells through the complexation of the T2-KK1B10 aptamer with the cells. The ATP in the cells and low level of intracellular Ca2+ ion release both the aptamer and ion from the surface of the MSNPs. The liberated rhodamine B results in an increased fluorescence intensity. Fluorescence microscope imaging and flow cytometry histogram display a strong fluorescence emission for the K562 cells (CML cells) exposed to the nanoconjugate in comparison with that for MCF-7 cells. The aptasensor possesses good performance in the blood samples with the advantages of high sensitivity, rapidness, and cost-effectiveness, making it an appropriate tool for the diagnosis of CML disease.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , Leucemia Mieloide , Humanos , Nanoconjugados/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/diagnóstico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Células K562 , Adenosina Trifosfato
3.
J Nanobiotechnology ; 21(1): 246, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37528408

RESUMEN

Type 2 diabetes mellitus (T2DM) predominantly considered a metabolic disease is now being considered an inflammatory disease as well due to the involvement of meta-inflammation. Obesity-induced adipose tissue inflammation (ATI) is one of the earliest phenomena in the case of meta-inflammation, leading to the advent of insulin resistance (IR) and T2DM. The key events of ATI are orchestrated by macrophages, which aggravate the inflammatory state in the tissue upon activation, ultimately leading to systemic chronic low-grade inflammation and Non-Alcoholic Steatohepatitis (NASH) through the involvement of proinflammatory cytokines. The CD44 receptor on macrophages is overexpressed in ATI, NASH, and IR. Therefore, we developed a CD44 targeted Hyaluronic Acid functionalized Graphene Oxide Quantum Dots (GOQD-HA) nanocomposite for tissue-specific delivery of metformin. Metformin-loaded GOQD-HA (GOQD-HA-Met) successfully downregulated the expression of proinflammatory cytokines and restored antioxidant status at lower doses than free metformin in both palmitic acid-induced RAW264.7 cells and diet induced obese mice. Our study revealed that the GOQD-HA nanocarrier enhanced the efficacy of Metformin primarily by acting as a therapeutic agent apart from being a drug delivery platform. The therapeutic properties of GOQD-HA stem from both HA and GOQD having anti-inflammatory and antioxidant properties respectively. This study unravels the function of GOQD-HA as a targeted drug delivery option for metformin in meta-inflammation where the nanocarrier itself acts as a therapeutic agent.


Asunto(s)
Diabetes Mellitus Tipo 2 , Metformina , Enfermedad del Hígado Graso no Alcohólico , Puntos Cuánticos , Animales , Ratones , Ácido Hialurónico/uso terapéutico , Puntos Cuánticos/uso terapéutico , Nanoconjugados/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Antioxidantes/uso terapéutico , Inflamación/tratamiento farmacológico , Citocinas , Metformina/farmacología , Metformina/uso terapéutico
4.
Front Immunol ; 14: 1212606, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37545526

RESUMEN

Background: The tumor microenvironment (TME) provides a conducive environment for the growth and survival of tumors. Negative factors present in TME, such as IL-10, may limit the effectiveness of cellular vaccines based on dendritic cells, therefore, it is important to control its effect. The influence of IL-10 on immune cells can be abolished e.g., by using antibodies against the receptor for this cytokine - anti-IL-10R. Furthermore, the anticancer activity of cellular vaccines can be enhanced by modifying them to produce proinflammatory cytokines, such as IL-12, IL-15 or IL-18. Additionally, an immunomodulatory dose of methotrexate and hydroxyethyl starch (HES-MTX) nanoconjugate may stimulate effector immune cells and eliminate regulatory T cells, which should enhance the antitumor action of immunotherapy based on DC vaccines. The main aim of our study was to determine whether the HES-MTX administered before immunotherapy with anti-IL-10R antibodies would change the effect of vaccines based on dendritic cells overproducing IL-12, IL-15, or IL-18. Methods: The activity of modified DCs was checked in two therapeutic protocols - immunotherapy with the addition of anti-IL10R antibodies and chemoimmunotherapy with HES-MTX and anti-IL10R antibodies. The inhibition of tumor growth and the effectiveness of the therapy in inducing a specific antitumor response were determined by analyzing lymphoid and myeloid cell populations in tumor nodules, and the activity of restimulated splenocytes. Results and conclusions: Using the HES-MTX nanoconjugate before immunotherapy based on multiple administrations of anti-IL-10R antibodies and cellular vaccines capable of overproducing proinflammatory cytokines IL-12, IL-15 or IL-18 created optimal conditions for the effective action of these vaccines in murine colon carcinoma MC38 model. The applied chemoimmunotherapy caused the highest inhibition of tumor growth in the group receiving DC/IL-15/IL-15Rα/TAg + DC/IL-18/TAg at the level of 72.4%. The use of cellular vaccines resulted in cytotoxic activity increase in both immuno- or chemoimmunotherapy. However, the greatest potential was observed both in tumor tissue and splenocytes obtained from mice receiving two- or three-component vaccines in the course of combined application. Thus, the designed treatment schedule may be promising in anticancer therapy.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias del Colon , Citocinas , Animales , Ratones , Células Dendríticas , Inmunoterapia/métodos , Interleucina-10 , Interleucina-12 , Interleucina-15 , Interleucina-18 , Metotrexato/farmacología , Metotrexato/uso terapéutico , Nanoconjugados/uso terapéutico , Microambiente Tumoral
5.
J Control Release ; 361: 636-658, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37544515

RESUMEN

Delivery of therapeutic substances into the brain poses a significant challenge in the treatment of neurological disorders. This is primarily due to the blood-brain barrier (BBB), which restricts access, alongside the limited stability and distribution of these agents within the brain tissue. Here we demonstrate an efficient delivery of microRNA (miRNA) and antisense RNA preferentially to neurons compared to astroglia in the brain of healthy and Alzheimer's disease mice, via disulfide-linked conjugation with poly(ß-L-malic acid-trileucine)-copolymer a biodegradable, amphiphilic, and multivalent platform. By conjugating a D-configured (D3)-peptide (vector) for specific targeting, highly efficient delivery across the BBB is achieved through the Low-Density Lipoprotein Receptor-Related Protein-1 (LRP-1) transcytosis pathway, amyloid beta (Aß) peptides. Nanodrug distribution was determined by fluorescent labeling and analyzed by microscopy in neurons, astroglia, and in extracellular amyloid plaques typical for Alzheimer's disease. Whereas D-configured BBB-vectors can efficiently target neurons, L-configured (e.g., AP2-peptide) guided vector can only cross BBB but not seem to bind neurons. An analysis of post-injection fluorescence distribution, and RNA-seq followed by real-time PCR validation, confirmed a successful in vivo delivery of morpholino-miRNA-186 nanoconjugates into mouse brain. The size and fluorescence intensity of the intracellular nanodrug particulates were analyzed and verified by a competition with non-fluorescent conjugates. Differentially expressed genes (DEGs) from RNA-seq were identified in the nanodrug injected mice, and the changes of selected DEGs related to Alzheimer's disease were further validated by western blot and real-time PCR. Collectively, these results demonstrated that D3-peptide-conjugated nanopolymer drug is able to achieve neuron-selective delivery of miRNA and can serve as an efficient brain delivery vehicle in Alzheimer's disease (AD) mouse models.


Asunto(s)
Enfermedad de Alzheimer , MicroARNs , Ácidos Nucleicos , Ratones , Animales , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/metabolismo , Ácidos Nucleicos/uso terapéutico , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Nanoconjugados/uso terapéutico , MicroARNs/uso terapéutico , Neuronas/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos
6.
Indian J Tuberc ; 70(3): 329-338, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37562909

RESUMEN

BACKGROUND: The biosynthesis of gold nanoparticles using medicinal plants as reducing and stabilizing agent for synthesis is an emerging area of research due to their cost effectiveness and further diversified applications in various fields. People with HIV are prone to these opportunistic infections like TB due to the immunocompromised condition. In the present study, the nanoparticles and nanoconjugates were screened for effective anti-mycobacterial efficiency against opportunistic infections. METHODS: Incidentally, the nanoparticles were biosynthesized using single plant extract. The biosynthesized nanoparticles were initially screened for effective anti-tuberculosis activity against Mycobacterium tuberculosis. Based on the effective antimicrobial activity, a nanoconjugate was biosynthesized combining three plant extracts for a cumulative activity. RESULTS: The biosynthesized gold nanoparticles and nanoconjugates showed MIC demonstrating for 99% inhibition and MIC99 was found to be 6.42 µg/ml. Among all the 15 nanoparticles tested, seven NPs showed exceptional anti-TB activities NP1, NP2, NP6, NP7, NP10, NP12 and NP15 and the other nanoparticles exhibited varying degrees of inhibition - anti-TB activities. In the 12 nanoconjugate tested, seven nanoconjugate demonstrated exceptional anti-TB activities such as NCC1, NCC2, NCC5, NCC6, NCV1, NCV6 and NCV4. CONCLUSION: The objective of the study was to identify the nanoparticles and nanoconjugates which demonstrated potential activity against M. tuberculosis so that a single nanoparticle or nanoconjugate can be targeted to treat patients with TB. Minimum Inhibitory Concentration (MIC) of the biosynthesized gold nanoparticles and nanoconjugates were determined against M. tuberculosis H37Rv.


Asunto(s)
Nanopartículas del Metal , Mycobacterium tuberculosis , Infecciones Oportunistas , Tuberculosis , Humanos , Nanoconjugados/uso terapéutico , Oro/farmacología , Oro/uso terapéutico , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Tuberculosis/tratamiento farmacológico , Infecciones Oportunistas/tratamiento farmacológico , Pruebas de Sensibilidad Microbiana
7.
Artif Cells Nanomed Biotechnol ; 51(1): 334-345, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37455406

RESUMEN

Metastatic melanoma cancer stem cells are subpopulations linked to tumour development, immunoevasive behaviour, treatment resistance, and metastasis, all of which contribute to a poor prognosis. Photodynamic treatment (PDT) is an alternate strategy to cancer eradication that involves the generation of reactive oxygen species. As a carrier, nanoparticles enable efficient cellular uptake of photosensitizers, improving organelle accumulation and cancer cell targeted therapy. This study considered at the effect of PDT on CD133+ Melanoma Stem Cells utilising an Aluminium Phthalocyanine Gold Nanoparticle (AlPcS4Cl-AuNP) combination. A ligand exchange approach was used to conjugate AlPcS4Cl-PEG-AuNP-COOH and was characterised using UV-Vis, FTIR, DLS and Zeta Potential. Stem cells isolated from the A375 cell line irradiated with a laser at 673.2 nm with a fluency of 5 J/cm2 were evaluated. Furthermore, it was important to study if apoptosis was one of the mechanisms causing to cell death which was substantiated with Annexin V/PI, caspase 3 and p53 analysis. The nanoparticle conjugate mediated PDT promoted apoptotic cell death, showing increased expression of p53 and caspase-3. The study proposed a strategy aimed at extending the understanding of PDT in enhancing the therapy of melanoma, suggesting a probable improved cell death when AlPcS4Cl was conjugated to AuNPs.


Asunto(s)
Melanoma , Nanopartículas del Metal , Fotoquimioterapia , Humanos , Nanoconjugados/uso terapéutico , Oro/metabolismo , Proteína p53 Supresora de Tumor , Línea Celular Tumoral , Melanoma/patología , Fármacos Fotosensibilizantes/farmacología , Apoptosis , Células Madre/metabolismo
8.
Front Immunol ; 14: 1155377, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033926

RESUMEN

Background: Understanding the negative impact of the tumor microenvironment on the creation of an effective immune response has contributed to the development of new therapeutic anti-cancer strategies. One such solution is combined therapy consisting of chemotherapeutic administration followed by dendritic cell (DC)-based vaccines. The use of cytostatic leads to the elimination of cancer cells, but can also modulate the tumor milieu. Moreover, great efforts are being made to increase the therapeutic outcome of immunotherapy, e.g. by enhancing the ability of DCs to generate an efficient immune response, even in the presence of immunosuppressive cytokines such as IL-10. The study aimed to determine the effectiveness of combined therapy with chemotherapeutic with immunomodulatory potential - HES-MTX nanoconjugate (composed of methotrexate (MTX) and hydroxyethyl starch (HES)) and DCs with downregulated expression of IL-10 receptor stimulated with tumor antigens (DC/shIL-10R/TAg) applied in MC38 murine colon carcinoma model. Methods: With the use of lentiviral vectors the DCs with decreased expression of IL-10R were obtained and characterized. During in vivo studies MC38-tumor bearing mice received MTX or HES-MTX nanoconjugate as a sole treatment or combined with DC-based immunotherapy containing unmodified DCs or DCs transduced with shRNA against IL-10R (or control shRNA sequence). Tumor volume was monitored during the experiment. One week after the last injection of DC-based vaccines, tumor nodules and spleens were dissected for ex vivo analysis. The changes in the local and systemic anti-tumor immune response were estimated with the use of flow cytometry and ELISA methods. Results and conclusions: In vitro studies showed that the downregulation of IL-10R expression in DCs enhances their ability to activate the specific anti-tumor immune response. The use of HES-MTX nanoconjugate and DC/shIL-10R/TAg in the therapy of MC38-tumor bearing mice resulted in the greatest tumor growth inhibition. At the local anti-tumor immune response level a decrease in the infiltration of cells with suppressor activity and an increase in the influx of effector cells into MC38 tumor tissue was observed. These changes were crucial to enhance the effective specific immune response at the systemic level, which was revealed in the greatest cytotoxic activity of spleen cells against MC38 cells.


Asunto(s)
Vacunas contra el Cáncer , Carcinoma , Neoplasias del Colon , Animales , Ratones , Metotrexato/farmacología , Metotrexato/uso terapéutico , Nanoconjugados/uso terapéutico , Microambiente Tumoral , ARN Interferente Pequeño/genética , Activación de Linfocitos , Células Dendríticas , Receptores de Interleucina-10/metabolismo , Carcinoma/tratamiento farmacológico
9.
Arch Microbiol ; 205(5): 170, 2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-37017767

RESUMEN

Balamuthia mandrillaris and Naegleria fowleri are protist pathogens that can cause fatal infections. Despite mortality rate of > 90%, there is no effective therapy. Treatment remains problematic involving repurposed drugs, e.g., azoles, amphotericin B and miltefosine but requires early diagnosis. In addition to drug discovery, modifying existing drugs using nanotechnology offers promise in the development of therapeutic interventions against these parasitic infections. Herein, various drugs conjugated with nanoparticles were developed and evaluated for their antiprotozoal activities. Characterizations of the drugs' formulations were accomplished utilizing Fourier-transform infrared spectroscopy, efficiency of drug entrapment, polydispersity index, zeta potential, size, and surface morphology. The nanoconjugates were tested against human cells to determine their toxicity in vitro. The majority of drug nanoconjugates exhibited amoebicidal effects against B. mandrillaris and N. fowleri. Amphotericin B-, Sulfamethoxazole-, Metronidazole-based nanoconjugates are of interest since they exhibited significant amoebicidal effects against both parasites (p < 0.05). Furthermore, Sulfamethoxazole and Naproxen significantly diminished host cell death caused by B. mandrillaris by up to 70% (p < 0.05), while Amphotericin B-, Sulfamethoxazole-, Metronidazole-based drug nanoconjugates showed the highest reduction in host cell death caused by N. fowleri by up to 80%. When tested alone, all of the drug nanoconjugates tested in this study showed limited toxic effects against human cells in vitro (less than 20%). Although these are promising findings, prospective work is warranted to comprehend the mechanistic details of nanoconjugates versus amoebae as well as their in vivo testing, to develop antimicrobials against the devastating infections caused by these parasites.


Asunto(s)
Amebiasis , Amebicidas , Balamuthia mandrillaris , Naegleria fowleri , Humanos , Anfotericina B/farmacología , Metronidazol/farmacología , Metronidazol/uso terapéutico , Nanoconjugados/química , Nanoconjugados/uso terapéutico , Estudios Prospectivos , Amebicidas/química , Amebicidas/farmacología , Sulfametoxazol/farmacología , Sulfametoxazol/uso terapéutico , Amebiasis/tratamiento farmacológico , Amebiasis/parasitología
10.
Drug Deliv ; 29(1): 1892-1902, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35748413

RESUMEN

The present study aimed to design and optimize, a nanoconjugate of gabapentin (GPN)-melittin (MLT) and to evaluate its healing activity in rat diabetic wounds. To explore the wound healing potency of GPN-MLT nanoconjugate, an in vivo study was carried out. Diabetic rats were subjected to excision wounds and received daily topical treatment with conventional formulations of GPN, MLT, GPN-MLT nanoconjugate and a marketed formula. The outcome of the in vivo study showed an expedited wound contraction in GPN-MLT-treated animals. This was confirmed histologically. The nanoconjugate formula exhibited antioxidant activities as evidenced by preventing malondialdehyde (MDA) accumulation and superoxide dismutase (SOD) and glutathione peroxidase (GPx) enzymatic exhaustion. Further, the nanoconjugate showed superior anti-inflammatory activity as it inhibited the expression of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). This is in addition to enhancement of proliferation as indicated by increased expression of transforming growth factor-ß (TGF- ß), vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor receptor-ß (PDGFRB). Also, nanoconjugate enhanced hydroxyproline concentration and mRNA expression of collagen type 1 alpha 1 (Col 1A1). In conclusion, a GPN-MLT nanoconjugate was optimized with respect to particle size. Analysis of pharmacokinetic attributes showed the mean particle size of optimized nanoconjugate as 156.9 nm. The nanoconjugate exhibited potent wound healing activities in diabetic rats. This, at least partly, involve enhanced antioxidant, anti-inflammatory, proliferative and pro-collagen activities. This may help to develop novel formulae that could accelerate wound healing in diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Factor A de Crecimiento Endotelial Vascular , Animales , Antiinflamatorios/uso terapéutico , Antioxidantes/metabolismo , Colágeno/metabolismo , Diabetes Mellitus Experimental/metabolismo , Gabapentina/metabolismo , Gabapentina/uso terapéutico , Meliteno/metabolismo , Meliteno/uso terapéutico , Nanoconjugados/uso terapéutico , Ratas , Ratas Wistar , Piel/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas
11.
J Appl Physiol (1985) ; 133(2): 262-272, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35771225

RESUMEN

Effects of the Adenosine A1 blockade using 8-cyclopentyl-1,3-diprophyxanthine (DPCPX) nanoconjugate on inducing recovery of the hemidiaphragm paralyzed by hemisection have been thoroughly examined previously; however, the toxicology of DPCPX nanoconjugate remains unknown. This research study investigates the therapeutic efficacy and toxicology of the nanoconjugate DPCPX in the cervical spinal cord injury (SCI) rat model. We hypothesized that a single injection of nanoconjugate DPCPX in the paralyzed left hemidiaphragm (LDH) of hemisected rats at the 2nd cervical segment (C2Hx) would lead to the long-term recovery of LDH while showing minimal toxicity. Adult male rats underwent left C2Hx surgery and the diaphragms' baseline electromyography (EMG). Subsequently, rats were randomized into a control group and four treated subgroups. Three subgroups received a single intradiaphragmatic dose of either 0.09, 0.15, or 0.27 µg/kg, and one subgroup received 0.1 mg/kg of native DPCPX two times per day intravenously (i.v.) for 3 days (total 0.6 mg/kg). Rats were monitored for a total of 56 days. Compared with control, the treatment with nanoconjugate DPCPX at 0.09 µg/kg, 0.15 µg/kg, and 0.27 µg/kg doses elicited significant recovery of paralyzed LDH (i.e., 67% recovery at 8 wk) (P < 0.05). DPCPX nanoconjugate-treated rats had significant weight loss for first 2 wk but recovered significantly by day 56 (P < 0.05). The levels of gold in the blood and body tissues were below the recommended levels. No sign of weakness, histology of tissue damage, or organ abnormality was observed. A dose of DPCPX nanoconjugate can induce long-term diaphragm recovery after SCI without observed toxicity.NEW & NOTEWORTHY The intradiaphragmatic administration of nanoconjugate is safe and has the promise to significantly reduce the therapeutic dosage for the treatment and achieve long-term and possibly permanent recovery in respiratory muscle dysfunction after SCI. No toxicity of nanoconjugate was found in any of the experimental animals.


Asunto(s)
Nanoconjugados , Traumatismos de la Médula Espinal , Xantinas , Animales , Diafragma , Masculino , Nanoconjugados/uso terapéutico , Nanoconjugados/toxicidad , Ratas , Recuperación de la Función , Traumatismos de la Médula Espinal/tratamiento farmacológico , Xantinas/uso terapéutico , Xantinas/toxicidad
12.
Int J Biol Macromol ; 210: 530-544, 2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35513094

RESUMEN

Among the most lethal forms of cancer, malignant brain tumors persist as one of the greatest challenges faced by oncologists, where nanotechnology-driven theranostics can play a critical role in developing novel polymer-based supramolecular nanoarchitectures with multifunctional and multi-modal characteristics to fight cancer. However, it is virtually a consensus that, besides the complexity of active delivering anticancer drugs by the nanocarriers to the tumor site, the current evaluation methods primarily relying on in vitro assays and in vivo animal models have been accounted for the low translational effectiveness to clinical applications. In this view, the chick chorioallantoic membrane (CAM) assay has been increasingly recognized as one of the best preclinical models to study the effects of anticancer drugs on the tumor microenvironment (TME). Thus, in this study, we designed, characterized, and developed novel hybrid nanostructures encompassing chemically functionalized carboxymethylcellulose (CMC) with mitochondria-targeting pro-apoptotic peptide (KLA) and cell-penetrating moiety (cysteine, CYS) with fluorescent inorganic semiconductor (Ag-In-S, AIS) for simultaneously bioimaging and inducing glioblastoma cancer cell (U-87 MG, GBM) death. The results demonstrated that the CMC-peptide macromolecules produced supramolecular vesicle-like nanostructures with aqueous colloidal stability suitable as nanocarriers for passive and active targeting of cancer tumors. The optical properties and physicochemical features of the nanoconjugates confirmed their suitability as photoluminescent nanoprobes for cell bioimaging and intracellular tracking. Moreover, the results in vitro demonstrated a notable killing activity towards GBM cells of cysteine-bearing CMC conjugates coupled with pro-apoptotic KLA peptides. More importantly, compared to doxorubicin (DOX), a model anticancer drug in chemotherapy that is highly toxic, these innovative nanohybrids nanoconjugates displayed higher lethality against U-87 MG cancer cells. In vivo CAM assays validated these findings where the nanohybrids demonstrated a significant reduction of GBM tumor progression (41% area) and evidenced an antiangiogenic activity. These results pave the way for developing polymer-based hybrid nanoarchitectonics applied as targeted multifunctional theranostics for simultaneous imaging and therapy against glioblastoma while possibly reducing the systemic toxicity and side-effects of conventional anticancer chemotherapeutic agents.


Asunto(s)
Antineoplásicos , Neoplasias Encefálicas , Glioblastoma , Puntos Cuánticos , Animales , Antineoplásicos/química , Neoplasias Encefálicas/tratamiento farmacológico , Carboximetilcelulosa de Sodio/química , Línea Celular Tumoral , Cisteína , Doxorrubicina/química , Glioblastoma/tratamiento farmacológico , Nanoconjugados/uso terapéutico , Polímeros/uso terapéutico , Puntos Cuánticos/química , Nanomedicina Teranóstica , Microambiente Tumoral
13.
Life Sci ; 298: 120525, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35378139

RESUMEN

AIMS: Synthesis of novel drug delivery system for targeted delivery of cuminaldehyde to breast cancer cells and the subsequent analyses of anti-neoplastic potential of the drug. MAIN METHODS: 3-carboxy-phenyl boronic acid (PBA) conjugated and polyacrylic acid (PAA) gated mesoporous silica nanoparticles (MSNs) were synthesized for the targeted delivery of cuminaldehyde (CUM) to breast cancer cells. Enhancement of anti-neoplastic effects of cuminaldehyde (4-isopropylbenzaldehyde) by the nanoconjugates was assessed. KEY FINDINGS: The anti-cancer effects of non-targeted and targeted drug-nanoconjugates were examined in vitro and in vivo. The targeted drug-nanoconjugates caused cell cycle arrest and induced the intrinsic pathway of apoptosis in MCF-7 cells through mitochondrial damage. In vivo intravenous injection of the targeted drug-nanoconjugates led to effective reduction in growth of 4 T1 induced mammary pad tumor in female BALB/c mice via augmented accumulation of cuminaldehyde. The drug-nanoconjugates did not exhibit any systemic toxicity. SIGNIFICANCE: Therefore, MSN-PBA-CUM-PAA represents a potent therapeutic model for breast cancer treatment.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Nanopartículas , Animales , Antineoplásicos/uso terapéutico , Benzaldehídos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Cimenos , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ratones , Nanoconjugados/uso terapéutico , Porosidad , Dióxido de Silicio/uso terapéutico
14.
PLoS One ; 17(2): e0264093, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35202419

RESUMEN

BACKGROUND: Lung cancer in men and women is considered the leading cause for cancer-related mortality worldwide. Anti-cancer peptides represent a potential untapped reservoir of effective cancer therapy. METHODOLOGY: Box-Behnken response surface design was applied for formulating Alendronate sodium (ALS)-mastoparan peptide (MP) nanoconjugates using Design-Expert software. The optimization process aimed at minimizing the size of the prepared ALS-MP nanoconjugates. ALS-MP nanoconjugates' particle size, encapsulation efficiency and the release profile were determined. Cytotoxicity, cell cycle, annexin V staining and caspase 3 analyses on A549 cells were carried out for the optimized formula. RESULTS: The results revealed that the optimized formula was of 134.91±5.1 nm particle size. The novel ALS-MP demonstrated the lowest IC50 (1.3 ± 0.34 µM) in comparison to ALS-Raw (37.6 ± 1.79 µM). Thus, the results indicated that when optimized ALS-MP nanoconjugate was used, the IC50 of ALS was also reduced by half. Cell cycle analysis demonstrated a significantly higher percentage of cells in the G2-M phase following the treatment with optimized ALS-MP nanoconjugates. CONCLUSION: The optimized ALS-MP formula had significantly improved the parameters related to the cytotoxic activity towards A549 cells, compared to control, MP and ALS-Raw.


Asunto(s)
Alendronato/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Nanoconjugados/uso terapéutico , Venenos de Avispas/farmacología , Células A549 , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Tamaño de la Partícula
15.
Curr Cancer Drug Targets ; 22(1): 31-48, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34872476

RESUMEN

Cancer is a rapidly growing life-threatening disease that affected 18.1 million people worldwide in 2018. Various conventional techniques like surgery, radiation, and chemotherapy are considered as a mainstream treatment for patients but show some limitations like cytotoxicity due to off-targeted action, poor intra-tumor localization, development of multi-drug resistance by tumor cells, physical and psychological stresses, etc. Such limitations have motivated the scientists to work towards more patient-centric and precision therapy using advanced drug delivery systems like liposomes, nanoparticles, nanoconjugates, etc. However, these carriers also face limitations like poor biocompatibility, lesser payload capacity, leakage of encapsulated drug, and short-term stability. So, this review article explores the profound insights for the development of biomacromolecule- functionalized nanoconjugates to potentiate the anticancer activity of therapeutic agents for various cancers like lung, colorectal, ovarian, breast and liver cancer. Researchers have shown interest in biofunctionalized nanoconjugates because of advantages like biocompatibility, site-specificity with better localization, higher entrapment with long-term stability and lesser off-target toxicity. The progressive trend of biomacromolecule nanoconjugates will encourage further research for the development of effective transport of drugs, nutraceuticals and phytoconstituents for on-site effect at cancer microenvironment and tumor cells with higher safety profile.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Antineoplásicos/uso terapéutico , Portadores de Fármacos , Sistemas de Liberación de Medicamentos/métodos , Humanos , Nanoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Microambiente Tumoral
16.
Med Mycol ; 60(2)2022 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-34958385

RESUMEN

Vulvovaginal candidiasis (VVC) is a commonly occurring yeast infection caused by Candida species in women. Among Candida species, C. albicans is the predominant member that causes vaginal candidiasis followed by Candida glabrata. Biofilm formation by Candida albicans on the vaginal mucosal tissue leads to VVC infection and is one of the factors for a commensal organism to get into virulent form leading to disease. In addition to that, morphological switching from yeast to hyphal form increases the risk of pathogenesis as it aids in tissue invasion. In this study, jacalin, a phytolectin complexed copper sulfide nanoparticles (NPs) have been explored to eradicate the mono and mixed species biofilms formed by fluconazole-resistant C. albicans and C. glabrata isolated from VVC patients. NPs along with standard antifungals like micafungin and amphotericin B have been evaluated to explore interaction behavior and we observed synergistic interactions between them. Microscopic techniques like light microscopy, phase contrast microscopy, scanning electron microscopy, confocal laser scanning microscopy were used to visualize the inhibition of biofilm by NPs and in synergistic combinations with standard antifungals. Real-time PCR analysis was carried out to study the expression pattern of the highly virulent genes which are responsible for yeast to hyphal switch, drug resistance and biofilm formation upon treatment with NPs in combination with standard antifungals. The current study shows that lectin-conjugated NPs with standard antifungals might be a different means to disrupt the mixed species population of Candida spp. that causes VVC. LAY SUMMARY: The present study focuses on exploiting the high biding affinity between the cell surface glycans present in Candida cells and the plant lectin, Jacalin. Jacalin serves as a 'Trojan Horse' wherein the lectin-coupled nanoparticles show a high efficacy when compared with the unconjugated nanoparticles. The present approach also improves the anti-biofilm activity of the antifungal drugs against drug-resistant Candida strains.


Asunto(s)
Candidiasis Vulvovaginal , Animales , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Biopelículas , Candida , Candida albicans , Candidiasis Vulvovaginal/tratamiento farmacológico , Candidiasis Vulvovaginal/veterinaria , Femenino , Pruebas de Sensibilidad Microbiana/veterinaria , Nanoconjugados/uso terapéutico , Virulencia
17.
Biomaterials ; 280: 121258, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34847435

RESUMEN

Current therapy in acute myeloid leukemia (AML) is based on chemotherapeutic drugs administered at high doses, lacking targeting selectivity and displaying poor therapeutic index because of severe adverse effects. Here, we develop a novel nanoconjugate that combines a self-assembled, multivalent protein nanoparticle, targeting the CXCR4 receptor, with an Oligo-Ara-C prodrug, a pentameric form of Ara-C, to highly increase the delivered payload to target cells. This 13.4 nm T22-GFP-H6-Ara-C nanoconjugate selectively eliminates CXCR4+ AML cells, which are protected by its anchoring to the bone marrow (BM) niche, being involved in AML progression and chemotherapy resistance. This nanoconjugate shows CXCR4-dependent internalization and antineoplastic activity in CXCR4+ AML cells in vitro. Moreover, repeated T22-GFP-H6-Ara-C administration selectively eliminates CXCR4+ leukemic cells in BM, spleen and liver. The leukemic dissemination blockage induced by T22-GFP-H6-Ara-C is significantly more potent than buffer or Oligo-Ara-C-treated mice, showing no associated on-target or off-target toxicity and, therefore, reaching a highly therapeutic window. In conclusion, T22-GFP-H6-Ara-C exploits its 11 ligands-multivalency to enhance target selectivity, while the Oligo-Ara-C prodrug multimeric form increases 5-fold its payload. This feature combination offers an alternative nanomedicine with higher activity and greater tolerability than current intensive or non-intensive chemotherapy for AML patients.


Asunto(s)
Antineoplásicos , Leucemia Mieloide Aguda , Profármacos , Animales , Antineoplásicos/farmacología , Citarabina/uso terapéutico , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Nanoconjugados/uso terapéutico , Profármacos/uso terapéutico
18.
Asian Pac J Cancer Prev ; 22(10): 3189-3201, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34710995

RESUMEN

OBJECTIVE: The present work was designed to study the effect of new conjugated caffeic and folic acid with silver nanoparticles with definite molecular size applied with and without gamma radiation exposure, as an antitumor agent against experimentally induced Ehrlich tumor and attempted to identify their potential molecular mechanisms of action throughout determination of anti-tumor activities using MTT cytotoxic assay against two human carcinoma cell lines in vitro, such as apoptosis analysis by flow cytometry through caspase-8, caspase-3 and TNF determination in vivo. MATERIALS AND METHODS: Adult female albino mice were used and divided into five groups. Animals were sacrificed and the following parameters were estimated, glutathione (GSH), glutathione peroxidase (GPx), superoxide dismutase (SOD) in blood in addition to caspase8, caspase 3 and tumor necrosis factor (TNF) of tumor tissue, liver and kidney function also measured in plasma. The tumor specimens were processed for histopathological examination. RESULTS: Nano-silver folate caffeic (NSFC) complex compound treatment resulted in growth inhibition in Ehrlich solid tumor, Hep-G2, and MCF-7 cells (IC50 0.062 mg, 7.70 µM, and 14.50 µM, respectively). Flow cytometric analysis revealed that (NSFC) with radiation IR had apoptotic effects at caspases 8 (Mean±SD) (49.4±14), caspase3 (39.97±9.75), and TNF (40.1±3.4) more than any other groups. Those disturbances were found to be associated with a kinetic induction of apoptosis and showed modulation of the antioxidant system {glutathione (GSH), glutathione peroxidase (GPx) and superoxide dismutase (SOD) which were 60.70±0.80, 26.73±0.80, 39.52±0.58 respectively}at the group which took (NSFC+IR), besides its high percentage of necrotic cells by histopathological studies. In conclusion, the present study showed that the treatment of (NSFC) exhibits very efficient oncolytic activity in delaying tumor growth in mice bearing Ehrlich Solid Carcinoma (ESC) and the mechanisms underlying the inhibitory effect of the present compound involve both an apoptotic effect against Hep-G2 and MCF-7 cells and modulation of antioxidant system.


Asunto(s)
Anticarcinógenos/uso terapéutico , Ácidos Cafeicos/uso terapéutico , Ácido Fólico/uso terapéutico , Nanopartículas del Metal/uso terapéutico , Neoplasias/prevención & control , Plata/uso terapéutico , Animales , Apoptosis , Neoplasias de la Mama/prevención & control , Carcinoma de Ehrlich , Caspasa 3/análisis , Caspasa 8/análisis , Femenino , Glutatión/sangre , Glutatión Peroxidasa/sangre , Células Hep G2 , Humanos , Neoplasias Hepáticas/prevención & control , Células MCF-7 , Ratones , Nanoconjugados/uso terapéutico , Neoplasias/patología , Superóxido Dismutasa/sangre , Factores de Necrosis Tumoral/análisis
19.
J Pharm Pharmacol ; 73(11): 1503-1512, 2021 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-34515769

RESUMEN

OBJECTIVES: The first goal of this study was to synthesize the silver nanoparticles Alcaligenes xylosoxidans exopolysaccharide (Ag-AXEPS). The second objective was to analyse the role of Ag-AXEPS nanoparticles (NPS) in treating bleomycin (BLM)-induced lung fibrosis. METHODS: Intratracheal bleomycin (2.5 U/kg) was administered to prompt pulmonary fibrosis in rats, and pulmonary fibrosis was treated with Ag-AXEPS nanoparticles (100 ppm/twice a week for four weeks). KEY FINDINGS: Ag-AXEPS nanoparticles significantly decreased the diversity of pulmonary inflammatory agents in rats with BLM-induced fibrosis. Reduced levels of respiratory tumor necrosis factor-alpha, monocyte chemotactic protein-1, matrix metalloproteinases (MMP-2 and MMP-9) were observed on treatment with synthesized Ag-AXEPS. Similarly, the treatment decreased IL-12, mRNA levels of BAX and plasma fibrosis markers like N-terminal procollagen III propeptide and transforming growth factor-ß1. On the other hand, the treatment increased mRNA BCL2 and total antioxidant capacity. It also lowered the level of fibrosis, as was shown by a quantified pathologic study of hematoxylin-eosin-stained lung parts. The treatment, however, ensured that lung collagen was restored, as assessed by Masson's trichrome stain, and that overall survival was increased and enhanced. CONCLUSIONS: Our work showed that nanoparticles could be obtained at 37°C and may be a possible pulmonary fibrosis therapeutic agent.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Bleomicina/efectos adversos , Pulmón/efectos de los fármacos , Nanopartículas/uso terapéutico , Polisacáridos Bacterianos/uso terapéutico , Fibrosis Pulmonar/tratamiento farmacológico , Plata/uso terapéutico , Alcaligenes , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Apoptosis , Colágeno/metabolismo , Fibrosis , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/prevención & control , Pulmón/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Nanopartículas del Metal/uso terapéutico , Nanoconjugados/uso terapéutico , Neumonía/inducido químicamente , Neumonía/metabolismo , Neumonía/prevención & control , Polisacáridos/farmacología , Polisacáridos/uso terapéutico , Polisacáridos Bacterianos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Ratas Sprague-Dawley , Plata/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Proteína X Asociada a bcl-2/metabolismo
20.
J Nanobiotechnology ; 19(1): 182, 2021 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-34127005

RESUMEN

BACKGROUND: Photodynamic therapy (PDT) may elicit antitumor immune response in addition to killing cancer cells. However, PDT as a monotherapy often fails to induce a strong immunity. Immune checkpoint inhibitors, which selectively block regulatory axes, may be used in combination with PDT to improve treatment outcomes. Indoleamine 2,3-dioxygenase (IDO) is an immunoregulatory enzyme and an important meditator of tumor immune escape. Combination therapy with PDT and IDO-targeted immune checkpoint blockage is promising but has been seldom been explored. METHODS: Herein we report a composite nanoparticle that allows for simultaneous delivery of photosensitizer and IDO inhibitor. Briefly, we separately load ZnF16Pc, a photosensitizer, and NLG919, an indoleamine 2,3-dioxygenase (IDO) inhibitor, into ferritin and poly(lactide-co-glycolic)-block-poly(ethylene glycol) (PEG-PLGA) nanoparticles; we then conjugate these two compartments to form a composite nanoparticle referred to as PPF NPs. We tested combination treatment with PPF NPs first in vitro and then in vivo in B16F10-tumor bearing C57/BL6 mice. RESULTS: Our results showed that PPF NPs can efficiently encapsulate both ZnF16Pc and NLG919. In vivo studies found that the combination treatment led to significantly improved tumor suppression and animal survival. Moreover, the treatment increased tumor infiltration of CD8+ T cells, while reducing frequencies of MDSCs and Tregs. 30% of the animals showed complete tumor eradication, and they successfully rejected a second tumor inoculation. Overall, our studies introduce a unique composite nanoplatform that allows for co-delivery of photosensitizer and IDO inhibitor with minimal inter-species interference, which is ideal for combination therapy.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Inmunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenasa/efectos de los fármacos , Nanoconjugados/uso terapéutico , Nanopartículas/uso terapéutico , Fotoquimioterapia/métodos , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Liberación de Fármacos , Inhibidores Enzimáticos/farmacología , Ferritinas , Humanos , Imidazoles , Isoindoles , Ratones , Células Supresoras de Origen Mieloide , Nanoconjugados/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...