Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Pharm ; 18(1): 87-100, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33231464

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC), a metabolic disorder, remains one of the leading cancer mortality sources worldwide. An initial response to treatments, such as gemcitabine (GEM), is often followed by emergent resistance reflecting an urgent need for alternate therapies. The PDAC resistance to GEM could be due to ERK1/2 activity. However, successful ERKi therapy is hindered due to low ligand efficiency, poor drug delivery, and toxicity. In this study, to overcome these limitations, we have designed pH-responsive nanoparticles (pHNPs) with a size range of 100-150 nm for the simultaneous delivery of ERKi (SCH 772984) and GEM with tolerable doses. These pHNPs are polyethylene glycol (PEG)-containing amphiphilic polycarbonate block copolymers with tertiary amine side chains. They are systemically stable and capable of improving in vitro and in vivo drug delivery at the cellular environment's acidic pH. The functional analysis indicates that the nanomolar doses of ERKi or GEM significantly decreased the 50% growth inhibition (IC50) of PDAC cells when encapsulated in pHNPs compared to free drugs. The combination of ERKi with GEM displayed a synergistic inhibitory effect. Unexpectedly, we uncover that the minimum effective dose of ERKi significantly promotes GEM activities on PDAC cells. Furthermore, we found that pHNP-encapsulated combination therapy of ERKi with GEM was superior to unencapsulated combination drug therapy. Our findings, thus, reveal a simple, yet efficient, drug delivery approach to overcome the limitations of ERKi for clinical applications and present a new model of sensitization of GEM by ERKi with no or minimal toxicity.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Portadores de Fármacos/química , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Nanopartículas/química , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/química , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Desnudos , Polietilenglicoles/química , Polímeros/química , Inhibidores de Proteínas Quinasas/química , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Gemcitabina
2.
Mol Pharm ; 17(12): 4676-4690, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33151075

RESUMEN

The molecular architecture of pH-responsive amphiphilic block copolymers, their self-assembly behavior to form nanoparticles (NPs), and doxorubicin (DOX)-loading technique govern the extent of DOX-induced cardiotoxicity. We observed that the choice of pH-sensitive tertiary amines, surface charge, and DOX-loading techniques within the self-assembled NPs strongly influence the release and stimulation of DOX-induced cardiotoxicity in primary cardiomyocytes. However, covalent conjugation of DOX to a pH-sensitive nanocarrier through a "conditionally unstable amide" linkage (PCPY-cDOX; PC = polycarbonate and PY = 2-pyrrolidine-1-yl-ethyl-amine) significantly reduced the cardiotoxicity of DOX in cardiomyocytes as compared to noncovalently encapsulated DOX NPs (PCPY-eDOX). When these formulations were tested for drug release in serum-containing media, the PCPY-cDOX systems showed prolonged control over drug release (for ∼72 h) at acidic pH compared to DOX-encapsulated nanocarriers, as expected. We found that DOX-encapsulated nanoformulations triggered cardiotoxicity in primary cardiomyocytes more acutely, while conjugated systems such as PCPY-cDOX prevented cardiotoxicity by disabling the nuclear entry of the drug. Using 2D and 3D (spheroid) cultures of an ER + breast cancer cell line (MCF-7) and a triple-negative breast cancer cell line (MDA-MB-231), we unravel that, similar to encapsulated systems (PCPY-eDOX-type) as reported earlier, the PCPY-cDOX system suppresses cellular proliferation in both cell lines and enhances trafficking through 3D spheroids of MDA-MB-231 cells. Collectively, our studies indicate that PCPY-cDOX is less cardiotoxic as compared to noncovalently encapsulated variants without compromising the chemotherapeutic properties of the drug. Thus, our studies suggest that the appropriate selection of the nanocarrier for DOX delivery may prove fruitful in shifting the balance between low cardiotoxicity and triggering the chemotherapeutic potency of DOX.


Asunto(s)
Cardiotoxicidad/prevención & control , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Neoplasias/tratamiento farmacológico , Polímeros/química , Animales , Animales Recién Nacidos , Cardiotoxicidad/etiología , Línea Celular Tumoral , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidad , Composición de Medicamentos/métodos , Liberación de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración de Iones de Hidrógeno , Miocitos Cardíacos , Nanopartículas/química , Neoplasias/patología , Cemento de Policarboxilato , Cultivo Primario de Células , Pirrolidinas/química , Ratas , Esferoides Celulares , Pruebas de Toxicidad Aguda
3.
ACS Appl Mater Interfaces ; 16(12): 14405-14420, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38490971

RESUMEN

Developing protein confinement platforms is an attractive research area that not only promotes protein delivery but also can result in artificial environment mimicking of the cellular one, impacting both the controlled release of proteins and the fundamental protein biophysics. Polymeric nanoparticles (PNPs) are attractive platforms to confine proteins due to their superior biocompatibility, low cytotoxicity, and controllable release under external stimuli. However, loading proteins into PNPs can be challenging due to the potential protein structural perturbation upon contacting the interior of PNPs. In this work, we developed a novel approach to encapsulate proteins in PNPs with the assistance of the zeolitic imidazolate framework (ZIF). Here, ZIF offers an additional protection layer to the target protein by forming the protein@ZIF composite via aqueous-phase cocrystallization. We demonstrated our platform using a model protein, lysozyme, and a widely studied PNP composed of poly(ethylene glycol)-poly(lactic-co-glycolic acid) (PEG-PLGA). A comprehensive study via standard loading and release tests as well as various spectroscopic techniques was carried out on lysozyme loaded onto PEG-PLGA with and without ZIF protection. As compared with the direct protein encapsulation, an additional layer with ZIF prior to loading offered enhanced loading capacity, reduced leaching, especially in the initial stage, led to slower release kinetics, and reduced secondary structural perturbation. Meanwhile, the function, cytotoxicity, and cellular uptake of proteins encapsulated within the ZIF-bound systems are decent. Our results demonstrated the use of ZIF in assisting in protein encapsulation in PNPs and established the basis for developing more sophisticated protein encapsulation platforms using a combination of materials of diverse molecular architectures and disciplines. As such, we anticipate that the protein-encapsulated ZIF systems will serve as future polymer protein confinement and delivery platforms for both fundamental biophysics and biochemistry research and biomedical applications where protein delivery is needed to support therapeutics and/or nutrients.


Asunto(s)
Estructuras Metalorgánicas , Nanopartículas , Poliésteres , Polietilenglicoles , Zeolitas , Estructuras Metalorgánicas/química , Muramidasa , Nanopartículas/química , Polímeros , Zeolitas/química
4.
ACS Appl Bio Mater ; 6(11): 4682-4693, 2023 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-37867293

RESUMEN

The three-dimensional (3D) spheroid cell culture model is crucial in screening anticancer drugs in vitro and understanding tumor cell behavior. However, the current in vitro models require highly skilled techniques. Here, we present an in vitro, tumor-mimetic, self-detachable, cancer cell spheroid model that provides the confined space of a tumor microenvironment, convenient spheroid retrieval, immunostaining, treatment, and imaging. We formed a void space within alginate macrobeads by ionic disintegration at a specific region inside. The macrobeads were further destabilized with bovine serum albumin to retrieve the spheroid cultured within the void space. Quantitative analysis of the immunofluorescence images of the cultured spheroids showed enhanced expressions of the hypoxia-inducible factor-1α (HIF-1α) and carbonic anhydrase-9 (CA-9), like monolayer cultures of cancer cells under hypoxic conditions (0.2% oxygen). Furthermore, adding CoCl2 to the cell culture media induces even higher amounts of HIF-1α and CA-9 in the cultured spheroids. In conclusion, the present work highlighted the in vitro spheroid model, which is closer to the tumor microenvironment and has user-friendly cell seeding, spheroid retrieval, and immunostaining steps.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Esferoides Celulares , Hidrogeles , Antineoplásicos/farmacología , Microambiente Tumoral
5.
ACS Biomater Sci Eng ; 9(5): 2103-2128, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-35679474

RESUMEN

Wearable, point-of-care diagnostics, and biosensors are on the verge of bringing transformative changes in detection, management, and treatment of cancer. Bioinspired materials with new forms and functions have frequently been used, in both translational and commercial spaces, to fabricate such diagnostic platforms. Engineered from organic or inorganic molecules, bioinspired systems are naturally equipped with biorecognition and stimuli-sensitive properties. Mechanisms of action of bioinspired materials are deeply connected with thermodynamically or kinetically controlled self-assembly at the molecular and supramolecular levels. Thus, integration of bioinspired materials into wearable devices, either as triggers or sensors, brings about unique device properties usable for detection, capture, or rapid readout for an analyte of interest. In this review, we present the basic principles and mechanisms of action of diagnostic devices engineered from bioinspired materials, describe current advances, and discuss future trends of the field, particularly in the context of cancer.


Asunto(s)
Materiales Biomiméticos , Técnicas Biosensibles , Neoplasias , Dispositivos Electrónicos Vestibles , Pruebas en el Punto de Atención , Neoplasias/diagnóstico
6.
Lab Chip ; 22(8): 1519-1530, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35266489

RESUMEN

Circulating tumor cells (CTCs) are distinct cancer biomarkers established in clinical settings for early cancer detection, metastasis progression, and minimal residual disease (MRD) monitoring. Despite numerous advances, the comprehensive molecular characterization of CTCs is extremely challenging owing to their rarity and heterogeneity. Here, we present a novel cotton microfluidic substrate (CMS) as an innovative biomedical matrix that efficiently isolates CTCs while facilitating in vitro CTC expansion to enable a further downstream analysis of these rare cells. CMS enabled static and dynamic isolation of cells from the MCF-7 cancer cell line, as well as from head and neck squamous cell carcinoma (HNSCC) patients' blood and the cell capture efficiencies were further compared with a clinically regulated OncoDiscover® Liquid Biopsy Test. Further, CMS acted as a matrix on which the captured cancer cells were grown in 3D tumor models for studying anti-cancer drug efficacy and multi-drug resistance (MDR) mechanisms. The design of the CMS employed two different surface chemistries, flattened and nanostructured surfaces, each conjugated to anti-EpCAM antibodies to evaluate the CTC capture efficiency and 3D tumor growth dynamics. The nanostructured surface was highly efficient for capturing CTCs and promoted 3D tumor spheroid formation with a 5-fold increase in size from day 03 to day 10 of culture. Moreover, when treated with an anti-cancer drug, cisplatin, an almost 1/2 reduction in tumor size was achieved within 24 hours, followed by a cytostatic threshold and eventual acquisition of drug resistance within 3 days. Conclusively, the CMS matrix exhibits potential for further development of "tissue on chip" and "point-of-care" medical devices in cancer diagnostics, and chemo-therapeutic efficacy evaluations in both drug discovery and development.


Asunto(s)
Antineoplásicos , Células Neoplásicas Circulantes , Anticuerpos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Separación Celular , Humanos , Biopsia Líquida , Células Neoplásicas Circulantes/patología
7.
Artículo en Inglés | MEDLINE | ID: mdl-35595620

RESUMEN

OBJECTIVE: The aim of this study was to investigate the presence of circulating tumor cells (CTCs) and their correlation with prognostic factors and clinical outcomes in treatment-naive patients with oral squamous cell carcinoma. STUDY DESIGN: CTCs were isolated using OncoDiscover technique from presurgically obtained peripheral blood of 152 patients with treatment naïve oral squamous cell carcinoma. Sensitivity analysis was performed by including 40 healthy controls. CTCs cutoff values for clinicopathologic factors were obtained from receiver operating characteristic curves. Multivariate models determined the significance of CTC as independent variables. Kaplan-Meier analysis differentiated in overall survival between CTC values corresponding to the stage. RESULTS: Sensitivity, specificity, and accuracy of CTC detection were 94.32%, 98%, and 95.17%, respectively. Platform differentiated true positives at >3.5 CTCs (P < .00001). CTCs above 20.5 were suggestive of nodal metastasis (P < .0001) with a linear trend for detecting occult metastasis (P = .061). Early and advanced stages could be differentiated by >13.5 CTCs (P < .0001). Elevated CTCs were significantly associated with extranodal extension (>21.45 CTCs, P = .025), perineural invasion (>19.35 CTCs, P = .049), and depth of invasion (>12.5 CTCs, P = .0038). Median survival was reduced by 19 months when CTCs were >13. CONCLUSIONS: Preoperative CTC levels demonstrated a strong correlation with adverse clinicopathology factors and suggested its role as a sensitive prognostic marker to predict survival outcome and disease progress.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Células Neoplásicas Circulantes , Biomarcadores de Tumor , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Humanos , Neoplasias de la Boca/terapia , Células Neoplásicas Circulantes/patología , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello
8.
Colloids Surf B Biointerfaces ; 200: 111563, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33454622

RESUMEN

New molecular motifs that can act as pH-regulating triggers for amphiphilic, pH-sensitive block copolymers are investigated. Inspired by the mechanism of action of pH-indicators, such as methyl orange, and natural amino acids, we designed these copolymers where either 4-Amino-4'-dimethylaminoazobenzene, AZB (pKa 3.4, an amine derivative of methyl orange), isoleucine, Ile (pKa 2.37 for carboxylic acid), or a statistical mixture of both were appended as side chains to the hydrophobic block to act as pH-triggers. These new side chain motifs were identified with an aim to enhance the self-assembling properties of the block copolymers in terms of particle size and stability, drug encapsulation, and release. As the parent polymer, poly (ethylene) glycol-block- poly (carbonate) (PEG-b-PC) of number average molecular weight 12.1 kDa was used. We observed that PEG-b-PC block copolymers, when engineered with AZB or Ile-type of pH-regulators appended as side chains to PC blocks, formed self-assembled, spherical nanoparticles with hydrodynamic diameters ranging from 114 to 137 nm depending on copolymer composition. Critical aggregation concentrations (CAC) of the block copolymers were found to be governed by the type and content of side chains. We explored the use of these newly designed block copolymer assemblies as drug carriers using gemcitabine (GEM) as a model cytotoxic drug generally used for pancreatic ductal adenocarcinoma (PDAC). We showed that AZB and Ile decorated copolymeric nanocarriers were able to encapsulate GEM at 13.8-28.8 % loading content and release the drug in a pH-dependent pattern. Drug-loaded nanocarriers showed cellular entry into PDAC cells in vitro and were found to exert cytotoxicity against these cells. Neither the block copolymers bearing AZB or Ile-type pH-responsive triggers, nor their self-assembled nanoparticles showed any cytotoxicity at usable concentrations, thereby reflecting the potentials of these molecular motifs for designing stimuli-responsive drug delivery nanosystems.


Asunto(s)
Sistemas de Liberación de Medicamentos , Polímeros , Portadores de Fármacos , Concentración de Iones de Hidrógeno , Micelas , Tamaño de la Partícula , Polietilenglicoles
9.
Nanoscale ; 13(41): 17473-17485, 2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34651162

RESUMEN

Designing nano-substrates (NS) that support three-dimensional (3D) cell growth using physico-chemical interventions mimicking the cellular microenvironment is highly challenging. Here we report NS that assist 3D cell development (3D NS) using multi-components on a glass substrate (2D GS), which mimics the ex vivo tissue microenvironment and promotes 3D cell growth superior to conventional 2D cell culturing methodologies. 3D NS were chemically fabricated by linking the combination of advanced materials imparting different physico-chemical traits, for example, multiwalled carbon nanotubes (CNT), graphene (G), bovine serum albumin (BSA), and iron oxide magnetic nanoparticles (MNP). We compared cell-substrate interactions resulting in cellular morphological changes, influence on the cell circularity index (CI), nuclear-cytoplasmic ratios (N/C), and nuclear compression or derangements using human colorectal carcinoma cells (HCT116) and cervical cancer (HeLa) cells. We observed the increase in N/C, extended on the 3D NS micro-environment as indicative of cellular adaptation and the transformation. HCT116 and HeLa cells on 2D GS showed an N/C ratio <0.3, and 3D NS cultured cells exhibited a higher N/C ratio (>0.5). The most significant increase in the ratio, relative to arrested cell spreading, was observed with G-3D NS. Furthermore, 3D NS were evaluated for the cell viability differentiations using the anticancer drug doxorubicin (Dox). The drug-treated cells on 3D NS demonstrated far-displaced N/C ratios compared to 2D GS. In conclusion, 3D NS systems implicate an 'in vitro to in vivo' relevance for the outcome in cell biology, cell proliferation and migration, and in anticancer drug efficacy evaluation.


Asunto(s)
Antineoplásicos , Nanotubos de Carbono , Antineoplásicos/farmacología , Proliferación Celular , Doxorrubicina/farmacología , Células HeLa , Humanos
10.
Nanoscale ; 13(44): 18799, 2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34747963

RESUMEN

Correction for 'Designing 3D-nanosubstrates mimicking biological cell growth: pitfalls of using 2D substrates in the evaluation of anticancer efficiency' by Ashwini Patil et al., Nanoscale, 2021, 13, 17473-17485, DOI: 10.1039/d1nr03816h.

11.
J Mater Chem B ; 9(13): 2946-2978, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33480960

RESUMEN

Advanced materials and chemo-specific designs at the nano/micrometer-scale have ensured revolutionary progress in next-generation clinically relevant technologies. For example, isolating a rare population of cells, like circulating tumor cells (CTCs) from the blood amongst billions of other blood cells, is one of the most complex scientific challenges in cancer diagnostics. The chemical tunability for achieving this degree of exceptional specificity for extra-cellular biomarker interactions demands the utility of advanced entities and multistep reactions both in solution and in the insoluble state. Thus, this review delineates the chemo-specific substrates, chemical methods, and structure-activity relationships (SARs) of chemical platforms used for isolation and enumeration of CTCs in advancing the relevance of liquid biopsy in cancer diagnostics and disease management. We highlight the synthesis of cell-specific, tumor biomarker-based, chemo-specific substrates utilizing functionalized linkers through chemistry-based conjugation strategies. The capacity of these nano/micro substrates to enhance the cell interaction specificity and efficiency with the targeted tumor cells is detailed. Furthermore, this review accounts for the importance of CTC capture and other downstream processes involving genotypic and phenotypic CTC analysis in real-time for the detection of the early onset of metastases progression and chemotherapy treatment response, and for monitoring progression free-survival (PFS), disease-free survival (DFS), and eventually overall survival (OS) in cancer patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patología , Humanos
12.
Bioelectricity ; 2(2): 158-166, 2020 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-32856018

RESUMEN

Background: Electrostatic complexes of poly (l-Arginine) (pArg) and hyaluronic acid (HA) have been investigated for their functional applications to supply free or polymeric form of l-Arginine (Arg) to target cells. As a vital amino acid, Arg plays significant role in multitude of pathophysiological processes ranging from wound healing to cancer. However, serum arginase expression and toxicity of Arg at cellular level renders exogenous delivery of this amino acid a challenging task. We showed that polyarginine-hyaluronic acid ionic nanocomplexes (pArg-HA iNCs) could be an effective way to deliver Arg to target cell populations. Materials and Methods: These electrostatic complexes were prepared by mixing HA (average m.w. of 200 kDa) with pArg (m.w. 5-15 kDa; Sigma) in aqueous solutions and purifying over glycerol. Nanocomplexes were characterized for their particle size, surface charge, capacity to release l-Arg, and intracellular uptake of complexes. Results: Synthesized nanocomplexes showed hydrodynamic diameter ranging from 140-306 nm depending on the content of pArg or HA within the formulation. With surface charge (ζ-potential) of -29 mV, the nanocomplexes showed pH-dependent release of Arg. At pH 7.4, pArg-HA iNCs released 30% of the total Arg-content, while at pH 5.0, 60% of Arg was released after 24 h. These electrostatically stabilized complexes were found to promote growth of human dermal fibroblasts (HDF) in wound-healing assay and increased nitric oxide (NO) activity in these cells in a time-dependent manner. Nanocomplexes also showed cellular uptake and enhanced dose-dependent toxicity against two pancreatic cancer cell lines, i.e. MIA PaCa-2 and Panc-1. Interestingly, the cytotoxic effect was synergized upon pre-treatment of the cells with a frontline chemotherapeutic agent, gemcitabine (GEM), and was not observed when the cells were treated with Arg alone. Conclusion: As such, this communication shows the prospect of pArg-HA iNC electrostatic nanocomplexes to interact and interfere with intracellular Arg metabolic machinery conducive to rescuing different pathological conditions.

13.
Biomater Sci ; 8(20): 5729-5738, 2020 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-32940277

RESUMEN

Here we report nanocarrier-anticancer drug conjugates culpable for cellular deformations, critically evidenced through image-based analysis as a measure of karyoplasmic ratio (KR) and nuclear surface area (NSA). Multiwalled carbon nanotubes (MWCNTs) were coordinated additionally with Fe3O4 nanoparticles (NPs) to evaluate the symbiotic influence, and further conjugated to Dox for evaluating the cellular kinetics and for measuring cell deformations. Cellular entry kinetics of the CNT (CNT-Dox and CNT-Cys-Fe3O4-Dox) nanocarriers and their efficiency in nuclear localization were evaluated using cervical cancer (HeLa) cells. Of note, the Dox-bound nanocarriers showed significantly enhanced cell toxicity over the free form of the drug. CNT-Dox and CNT-Cys-Fe3O4-Dox influx occurred within 4 hours, while maximum cellular retention of Dox was observed for CNT-Dox at 24 h. However, the highest KR (∼0.51) was observed for CNT-Dox within 8 hours indicating similar cellular deformations using nanocarrier anticancer drug-conjugates to that of free Dox (KR ∼0.50) at 4 hours. In addition, we observed increased NSA at 4 h in Dox treatment whereas in the case of the Dox conjugated nanocarrier, increased NSA was noted at 8 h treatment. At 8 h exposure of HeLa cells with Dox conjugates, we observed that the cells fall into distinct regions of the morphospace with respect to KR and NSA. Conclusively, nano delivery systems considered for clinical and biomedical translations must take into account the possible negative influences imparting higher cellular deformations and secondary adverse effects over the free form of the drug.


Asunto(s)
Antineoplásicos , Nanopartículas , Nanotubos de Carbono , Antineoplásicos/uso terapéutico , Doxorrubicina , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Células HeLa , Humanos
14.
Nanoscale Adv ; 2(12): 5746-5759, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36133866

RESUMEN

Control of the sub-cellular localization of nanoparticles (NPs) with enhanced drug-loading capacity, employing graphene oxide (GO), iron oxide (Fe3O4) NPs and sandwiched deoxyribonucleic acid (DNA) bearing intercalated anticancer drug doxorubicin (DOX) has been investigated in this work. The nanosystems G-DNA-DOX-Fe3O4 and Fe3O4-DNA-DOX differentially influence serum protein binding and deliver DOX to lysosomal compartments of cervical cancer (HeLa) cells with enhanced retention. Stern-Volmer plots describing BSA adsorption on the nanosystems demonstrated the quenching constants, K sv for G-DNA-DOX-Fe3O4 and Fe3O4-DNA-DOX (0.025 mL µg-1 and 0.0103 mL µg-1 respectively). Nuclear DOX intensity, measured at 24 h, was ∼2.0 fold higher for Fe3O4-DNA-DOX in HeLa cells. Parallelly, the cytosol displayed ∼2.2 fold higher DOX intensity for Fe3O4-DNA-DOX compared to G-DNA-DOX-Fe3O4. Fe3O4-DNA-DOX was more efficacious in the cytotoxic effect than G-DNA-DOX-Fe3O4 (viability of treated cells: 33% and 49% respectively). The DNA:nanosystems demonstrated superior cytotoxicity compared to mole-equivalent free DOX administration. The results implicate DNA:DOX NPs in influencing the cellular uptake mechanism and were critically subject to cellular localization. Furthermore, cell morphology analysis evidenced maximum deformation attributed to free-DOX with 34% increased cell roundness, 63% decreased cell area and ∼1.9 times increased nuclear-to-cytoplasmic (N/C) ratio after 24 h. In the case of Fe3O4-DNA-DOX, the N/C ratio increased 1.2 times and a maximum ∼37% decrease in NSA was noted suggesting involvement of non-canonical cytotoxic pathways. In conclusion, the study makes a case for designing nanosystems with controlled and regulated sub-cellular localization to potentially exploit secondary cytotoxic pathways, in addition to optimized drug-loading for enhanced anticancer efficacy and reduced adverse effects.

15.
Sci Rep ; 10(1): 16607, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33004848

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

16.
Nanoscale Adv ; 2(6): 2315-2325, 2020 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-36133387

RESUMEN

Spatial arrangement for cells and the opportunity thereof have implications in cell regeneration and cell proliferation. 3D inverse space (3DIS) substrates with micron-sized pores are fabricated under controlled environmental conditions from polymers such as poly(lactic-co-glycolic) acid (PLGA), poly(lactic acid) (PLA) and poly(styrene) (PS). The characterization of 3DIS substrates by optical microscopy, scanning probe microscopy (SPM), etc. shows pores within 1-18 µm diameter and prominent surface roughness extending up to 3.9 nm in height over its base. Conversely, to compare two-dimensional (2D) versus 3DIS substrates, the crucial variables of cell height, cell spreading area and cell volume are compared using lung adenocarcinoma (A549) cells. The results indicate an average cell thickness of ∼6 µm on a glass substrate whereas cells on PLGA 3DIS were ∼12 µm in height, occasionally reaching 20 µm, with a 40% decreased cell spreading area. A549 cells cultured on polymer 3DIS substrates show a cell regeneration growth pattern, dependent on the available spatial volume. Furthermore, PLGA 3DIS cell culture systems with and without graded doxorubicin (DOX) pre-treatment result in potent cell inhibition and cell proliferation, respectively. Additionally, standard DOX administration to A549 cells in the PLGA 3DIS system revealed altered drug sensitivity. 3DIS demonstrates utility in facilitating cellular regeneration and mimicking cell proliferation in defined spatial arrangements.

17.
Sci Rep ; 10(1): 10010, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32561829

RESUMEN

Herein we report a hierarchically organized, water-dispersible 'nanocage' composed of cellulose nanocrystals (CNCs), which are magnetically powered by iron oxide (Fe3O4) nanoparticles (NPs) to capture circulating tumor cells (CTCs) in blood for head and neck cancer (HNC) patients. Capturing CTCs from peripheral blood is extremely challenging due to their low abundance and its account is clinically validated in progression-free survival of patients with HNC. Engaging multiple hydroxyl groups along the molecular backbone of CNC, we co-ordinated Fe3O4 NPs onto CNC scaffold, which was further modified by conjugation with a protein - transferrin (Tf) for targeted capture of CTCs. Owing to the presence of Fe3O4 nanoparticles, these nanocages were magnetic in nature, and CTCs could be captured under the influence of a magnetic field. Tf-CNC-based nanocages were evaluated using HNC patients' blood sample and compared for the CTC capturing efficiency with clinically relevant Oncoviu platform. Conclusively, we observed that CNC-derived nanocages efficiently isolated CTCs from patient's blood at 85% of cell capture efficiency to that of the standard platform. Capture efficiency was found to vary with the concentration of Tf and Fe3O4 nanoparticles immobilized onto the CNC scaffold. We envision that, Tf-CNC platform has immense connotation in 'liquid biopsy' for isolation and enumeration of CTCs for early detection of metastasis in cancer.


Asunto(s)
Celulosa , Neoplasias de Cabeza y Cuello/patología , Nanopartículas , Células Neoplásicas Circulantes/patología , Transferrina , Separación Celular , Humanos
18.
J Mater Chem B ; 8(9): 1852-1862, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32022091

RESUMEN

Cancer diagnosis and its stage-wise assessment are determined through invasive solid tissue biopsies. Conversely, cancer imaging is enriched through emission tomography and longitudinal high-resolution analysis for the early detection of cancer through altered cell morphology and cell-deformation. Similarly, in post multiple chemo-cycle exposures, the tumor regression and progression thereafter are not well understood. Here, we report chemo-cycles of doxorubicin (Dox) carrying nanoparticles (NPs) to be highly indicative of cell deformation and a progressive indicator of phenotypic expressions of acquired drug resistance (ADR). We designed graphene (G) based nanocarriers by chemically conjugating multiple components: (i) G; (ii) iron oxide (Fe3O4) NPs; and (iii) Dox through a cysteine (Cys) linker (G-Dox and G-Cys-Fe3O4-Dox). Although Dox underwent cell diffusion, the G-based nanocarriers followed a receptor-mediated endocytosis which created a profound impact on the cell membrane integrity. ADR owing to Dox and G-based nanocarriers was analyzed through a cytotoxicity assay, cell morphology deformation parameters and cellular uptake kinetic patterns. Interestingly, after the third chemo-cycle, G-Dox incubated cells showed the greatest decrease in the alteration of the nuclear surface area (NSA) of ∼28%, a ∼40% reduction of the cell surface area (CSA) and a ∼32% increase in the cell roundness (CRd). Our results suggested that the G-based nanocarriers induced the cell deformation process, subsequently resulting in ADR. Although the G-based nanocarriers initiated ADR, G-Dox was most cytotoxic to cancer cells and induced the maximum cell morphology deformation within our scope of study. This outcome implies caution is needed when using G-based nanocarriers and other multi-component nanosystems for Dox delivery as they lead to possible phenotypic expressions of drug resistance in cancer cells.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Grafito/química , Nanopartículas/química , Antibióticos Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/química , Portadores de Fármacos/química , Resistencia a Antineoplásicos/efectos de los fármacos , Células HeLa , Humanos , Tamaño de la Partícula , Propiedades de Superficie
19.
Biomater Sci ; 8(18): 4887-4905, 2020 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-32830832

RESUMEN

Electrospinning is historically related to tissue engineering due to its ability to produce nano-/microscale fibrous materials with mechanical and functional properties that are extremely similar to those of the extracellular matrix of living tissues. The general interest in electrospun fibrous matrices has recently expanded to cancer research both as scaffolds for in vitro cancer modelling and as patches for in vivo therapeutic delivery. In this review, we examine electrospinning by providing a brief description of the process and overview of most materials used in this process, discussing the effect of changing the process parameters on fiber conformations and assemblies. Then, we describe two different applications of electrospinning in service of cancer research: firstly, as three-dimensional (3D) fibrous materials for generating in vitro pre-clinical cancer models; and secondly, as patches encapsulating anticancer agents for in vivo delivery.


Asunto(s)
Nanofibras , Neoplasias , Matriz Extracelular , Neoplasias/tratamiento farmacológico , Ingeniería de Tejidos , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA