Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Asian J Pharm Sci ; 19(1): 100858, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38362469

ABSTRACT

Hydrogen sulfide (H2S) is a toxic, essential gas used in various biological and physical processes and has been the subject of many targeted studies on its role as a new gas transmitter. These studies have mainly focused on the production and pharmacological side effects caused by H2S. Therefore, effective strategies to remove H2S has become a key research topic. Furthermore, the development of novel nanoplatforms has provided new tools for the targeted removal of H2S. This paper was performed to review the association between H2S and disease, related H2S inhibitory drugs, as well as H2S responsive nanoplatforms (HRNs). This review first analyzed the role of H2S in multiple tissues and conditions. Second, common drugs used to eliminate H2S, as well as their potential for combination with anticancer agents, were summarized. Not only the existing studies on HRNs, but also the inhibition H2S combined with different therapeutic methods were both sorted out in this review. Furthermore, this review provided in-depth analysis of the potential of HRNs about treatment or detection in detail. Finally, potential challenges of HRNs were proposed. This study demonstrates the excellent potential of HRNs for biomedical applications.

2.
J Control Release ; 367: 167-183, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37562556

ABSTRACT

The tumor microenvironment is a barrier to breast cancer therapy. Cancer-associated fibroblast cells (CAFs) can support tumor proliferation, metastasis, and drug resistance by secreting various cytokines and growth factors. Abnormal angiogenesis provides sufficient nutrients for tumor proliferation. Considering that CAFs express the sigma receptor (which recognizes anisamide, AA), we developed a CAFs and breast cancer cells dual-targeting nano drug delivery system to transport the LightOn gene express system, a spatiotemporal controlled gene expression consisting of a light-sensitive transcription factor and a specific minimal promoter. We adopted RGD (Arg-Gly-Asp) to selectively bind to the αvß3 integrin on activated vascular endothelial cells and tumor cells. After the LightOn system has reached the tumor site, LightOn gene express system can spatiotemporal controllably express toxic Pseudomonas exotoxin An under blue light irradiation. The LightOn gene express system, combined with multifunctional nanoparticles, achieved high targeting delivery efficiency both in vitro and in vivo. It also displayed strong tumor and CAFs inhibition, anti-angiogenesis ability and anti-metastasis ability, with good safety. Moreover, it improved survival rate, survival time, and lung metastasis rate in a mouse breast cancer model. This study proves the efficacy of combining the LightOn system with targeted multifunctional nanoparticles in tumor and anti-metastatic therapy and provides new insights into tumor microenvironment regulation.


Subject(s)
Multifunctional Nanoparticles , Nanoparticles , Neoplasms , Mice , Animals , Endothelial Cells , Exotoxins/genetics , Exotoxins/therapeutic use , Gene Expression Regulation , Transgenes , Cell Line, Tumor , Tumor Microenvironment , Nanoparticles/therapeutic use
3.
ACS Appl Mater Interfaces ; 15(46): 53198-53216, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37942626

ABSTRACT

The increased risk of breast cancer metastasis is closely linked to the effects of platelets. Our previously light-switchable diphtheria toxin A fragment (DTA) gene system, known as the LightOn system, has demonstrated significant therapeutic potential; it lacks antimetastatic capabilities. In this study, we devised an innovative system by combining cell membrane fusion liposomes (CML) loaded with the light-switchable transgene DTA (pDTA) and a ticagrelor (Tig) prodrug. This innovative system, named the sequential rocket-mode bioactivating drug delivery system (pDTA-Tig@CML), aims to achieve targeted pDTA delivery while concurrently inhibiting platelet activity through the sequential release of Tig triggered by reactive oxygen species with the tumor microenvironment. In vitro investigations have indicated that pDTA-Tig@CML, with its ability to sequentially release Tig and pDTA, effectively suppresses platelet activity, resulting in improved therapeutic outcomes and the mitigation of platelet driven metastasis in breast cancer. Furthermore, pDTA-Tig@CML exhibits enhanced tumor aggregation and successfully restrains tumor growth and metastasis. It also reduces the levels of ADP, ATP, TGF-ß, and P-selectin both in vitro and in vivo, underscoring the advantages of combining the bioactivating Tig prodrug nanoplatform with the LightOn system. Consequently, pDTA-Tig@CML emerges as a promising light-switchable DTA transgene system, offering a novel bioactivating prodrug platform for breast cancer treatment.


Subject(s)
Breast Neoplasms , Prodrugs , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Prodrugs/pharmacology , Prodrugs/therapeutic use , Ticagrelor/pharmacology , Cell Line, Tumor , Liposomes , Transgenes , Tumor Microenvironment , Melanoma, Cutaneous Malignant
4.
Int J Pharm ; 644: 123249, 2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37467816

ABSTRACT

Breast cancer, which requires comprehensive multifunctional treatment strategies, is a major threat to the health of women. To develop multifunctional treatment strategies, we combined photothermal therapy (PTT) with immunotherapy in multifunctional nanoparticles for enhancing the anti-tumor efficacy. Fe3O4 nanoparticles coated with the polydopamine shell modified with polyethylene glycol and cyclic arginine-glycyl-aspartic peptide/anisamide (tNP) for loading the immune adjuvant resiquimod (R848) (R848@tNP) were developed in this research. R848@tNP had a round-like morphology with a mean diameter of 174.7 ± 3.8 nm, the zeta potential of -20.9 ± 0.9 mV, the drug loading rate of 9.2 ± 1.1 %, the encapsulation efficiency of 81.7 ± 3.2 %, high photothermal conversion efficiency and excellent magnetic properties in vitro. Furthermore, this research also explored the anticancer efficacy of nanoparticles against the breast cancer under the near-infrared (NIR) light (808 nm) in vitro and in vivo. R848@tNP-based NIR therapy effectively inhibited the proliferation of breast cancer cells. Moreover, R848@tNP mediated PTT significantly enhanced the maturation of dendritic cells in vitro. Additionally, R848@tNP enhances the anti-tumor effect and evoked an immune response under NIR in vivo. Furthermore, the biosafety of R848@tNP was fully investigated in this study. Collectively, these results clearly demonstrate that R848@tNP, with magnetic resonance imaging characteristics, is a potential therapeutic for breast cancer that combines PTT with the immunotherapy.


Subject(s)
Breast Neoplasms , Nanoparticles , Female , Humans , Breast Neoplasms/drug therapy , Phototherapy , Immunotherapy
5.
J Control Release ; 355: 538-551, 2023 03.
Article in English | MEDLINE | ID: mdl-36063962

ABSTRACT

Individualized immunotherapy has attracted great attention due to its high specificity, effectiveness, and safety. We used an exogenous antigen to label tumor cells with MHC I molecules, which allowed neoantigen-specific T cells to recognize and kill tumor cells. A neoantigen vaccine alone cannot achieve complete tumor clearance due to a tumor immunosuppressive microenvironment. The LightOn system was developed to effectively eliminate tumor cells through the spatiotemporally controllable expression of diphtheria toxin A fragment, leading to antigen release in the tumor region. These antigens stimulated and enhanced immunological function and thus, recruited neoantigen-specific T cells to infiltrate tumor tissue. Using the nanoparticle delivery system, neoantigens produced higher delivery efficiency to lymph nodes and improved tumor targeting ability for tumor cell labelling. Good tumor inhibition and prolonged survival were achieved, while eliciting a strong immune response. The combination of a spatiotemporally controllable transgene system with tumor neoantigen labeling has great potential for tumor immunotherapy.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Antigens, Neoplasm , Neoplasms/therapy , T-Lymphocytes , Immunotherapy , Histocompatibility Antigens Class I , Cancer Vaccines/genetics , Tumor Microenvironment
6.
Int J Pharm ; 624: 122018, 2022 Aug 25.
Article in English | MEDLINE | ID: mdl-35839982

ABSTRACT

Photodynamic therapy (PDT) induces tumour cell death by producing reactive oxygen species (ROS), and hypoxia is one of the main factors that limits its efficiency. In our previous study, bufalin (BU) enhanced photosensitizer mTHPC-mediated PDT therapy in colorectal cancer (CRC) cells, but its mechanism was not elucidated. To explore a strategy for improving the efficacy of PDT, we designed iRGD-modified nanoparticles to co-capsuled mTHPC and BU for simultaneous delivery to the tumour site and explored the underlying mechanism of the synergistic anti-CRC effect. In our study, mTHPC&BU@VES-CSO/TPGS-RGD nanoparticles (T-B@NP) had a particle size of 148.3 ± 2.5 nm and a zeta potential of 22.8 ± 2.0 mV. Specifically, these nanoparticles passively accumulated in tumour cells, and under laser irradiation, mTHPC induced cell apoptosis and death. In addition, the sustained release of BU inhibited HIF-1α and reduced VEGF-mediated angiogenesis by targeting the SRC-3/HIF-1α pathway, which induced a strong PDT effect against CRC. In vivo studies demonstrated that codelivery of the nanoparticles under laser irradiation exhibited a superior antitumour effect (84.2%) and significantly prolonged survival time of mice, with the mechanisms of alleviating hypoxia and inhibiting angiogenesis. In summary, mTHPC and BU codelivery via nanoparticles efficiently enhances the therapeutic effects of PDT by inhibiting the SRC-3/HIF-1α pathway in CRC. This work provides an effective strategy to combat hypoxia-induced tumour resistance and overcome the barriers of PDT treatment.


Subject(s)
Colorectal Neoplasms , Photochemotherapy , Animals , Bufanolides , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Hypoxia/drug therapy , Mice , Neovascularization, Pathologic/drug therapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use
7.
J Transl Med ; 19(1): 383, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34496870

ABSTRACT

BACKGROUND: Antiangiogenic therapy has increasingly become an important strategy for the treatment of colorectal cancer. Recent studies have shown that the tumour microenvironment (TME) promotes tumour angiogenesis. Bufalin is an active antitumour compound whose efficacy has been indicated by previous studies. However, there are very few studies on the antiangiogenic effects of bufalin. METHODS: Herein, human umbilical vein endothelial cell (HUVEC) tube formation, migration and adhesion tests were used to assess angiogenesis in vitro. Western blotting and quantitative PCR were used to detect relevant protein levels and mRNA expression levels. A subcutaneous xenograft tumour model and a hepatic metastasis model were established in mice to investigate the influence of bufalin on angiogenesis mediated by the TME in vivo. RESULTS: We found that angiogenesis mediated by cells in the TME was significantly inhibited in the presence of bufalin. The results demonstrated that the proangiogenic genes in HUVECs, such as VEGF, PDGFA, E-selectin and P-selectin, were downregulated by bufalin and that this downregulation was mediated by inhibition of the STAT3 pathway. Overexpression of STAT3 reversed the inhibitory effects of bufalin on angiogenesis. Furthermore, there was little reduction in angiogenesis when bufalin directly acted on the cells in the tumour microenvironment. CONCLUSION: Our findings demonstrate that bufalin suppresses tumour microenvironment-mediated angiogenesis by inhibiting the STAT3 signalling pathway in vascular endothelial cells, revealing that bufalin may be used as a new antiangiogenic adjuvant therapy medicine to treat colorectal cancer.


Subject(s)
Liver Neoplasms , Tumor Microenvironment , Angiogenesis Inhibitors/pharmacology , Animals , Bufanolides , Cell Movement , Cell Proliferation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Liver Neoplasms/drug therapy , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , STAT3 Transcription Factor/metabolism
8.
Mol Ther ; 29(10): 2931-2948, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34023507

ABSTRACT

Checkpoint inhibitors, such as anti-PD-1/PD-L1 antibodies, have been shown to be extraordinarily effective, but their durable response rate remains low, especially in colorectal cancer (CRC). Recent studies have shown that photodynamic therapy (PDT) could effectively enhance PD-L1 blockade therapeutic effects, although the reason is still unclear. Here, we report the use of multifunctional nanoparticles (NPs) loaded with photosensitized mTHPC (mTHPC@VeC/T-RGD NPs)-mediated PDT treatment to potentiate the anti-tumor efficacy of PD-L1 blockade for CRC treatment and investigate the underlying mechanisms of PDT enhancing PD-L1 blockade therapeutic effect in this combination therapy. In this study, the mTHPC@VeC/T-RGD NPs under the 660-nm near infrared (NIR) laser could kill tumor cells by inducing apoptosis and/or necrosis and stimulating systemic immune response, which could be further promoted by the PD-L1 blockade to inhibit primary and distant tumor growth, as well as building long-term host immunological memory to prevent tumor recurrence. Furthermore, we detected that mTHPC@VeC/T-RGD NP-mediated PDT sensitizes tumors to PD-L1 blockade therapy mainly because PDT-mediated hypoxia could induce the hypoxia-inducible factor 1α (HIF-1α) signaling pathway that upregulates PD-L1 expression in CRC. Taken together, our work demonstrates that mTHPC@VeC/T-RGD NP-mediated PDT is a promising strategy that may potentiate the response rate of anti-PD-L1 checkpoint blockade immunotherapies in CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immune Checkpoint Inhibitors/administration & dosage , Photochemotherapy/methods , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Immune Checkpoint Inhibitors/pharmacology , Mesoporphyrins/chemistry , Mesoporphyrins/pharmacology , Mice , Multifunctional Nanoparticles/administration & dosage , Multifunctional Nanoparticles/chemistry , Particle Size , Tumor Hypoxia/drug effects
9.
Cancer Lett ; 513: 63-74, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34000344

ABSTRACT

M2-polarized macrophages are one of critical factors in tumour chemoresistance. An increasing number of studies have shown that M2 macrophage polarization can be promoted by chemoresistance. A large number of evidences indicate that Bufalin has significant antitumour effect, previous studies have found that Bufalin can reduce the polarization of M2 macrophages to play an anti-tumour effect in vivo, but the mechanism remains unclear. In our study, we found that Bufalin reduced the polarization of M2 macrophages induced by chemoresistant cells both in vivo and in vitro; however, Bufalin had no obvious direct effect on M2 macrophage polarization. Furthermore, we demonstrated that Bufalin targeted the SRC-3 protein to reduce MIF release in chemoresistant cells in order to regulate the polarization of M2 macrophages. More interestingly, we also found that Cinobufacini, Bufalin is its main active monomer, which its could regulate the polarization of M2 macrophages to enhance the anti-tumour effect of oxaliplatin in vivo and in the clinic. Overall, this study provides a theoretical basis for the clinical application of drugs containing Bufalin as the main active ingredient in combination with established chemotherapy for the treatment of colorectal cancer.


Subject(s)
Bufanolides/therapeutic use , Colorectal Neoplasms/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/metabolism , Sodium-Potassium-Exchanging ATPase/therapeutic use , Animals , Bufanolides/pharmacology , Colorectal Neoplasms/pathology , Humans , Mice , Sodium-Potassium-Exchanging ATPase/pharmacology
10.
Acta Pharm Sin B ; 10(9): 1741-1753, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33088693

ABSTRACT

A light-switchable transgene system called LightOn gene expression system could regulate gene expression with a high on/off ratio under blue light, and have great potential for spatiotemporally controllable gene expression. We developed a nanoparticle drug delivery system (NDDS) to achieve tumor microenvironment-responsive and targeted delivery of diphtheria toxin A (DTA) fragment-encoded plasmids to tumor sites. The expression of DTA was induced by exposure to blue light. Nanoparticles composed of polyethylenimine and vitamin E succinate linked by a disulfide bond, and PEGylated hyaluronic acid modified with RGD peptide, accumulated in tumor tissues and were actively internalized into 4T1 cells via dual targeting to CD44 and α v ß 3 receptors. The LightOn gene expression system was able to control target protein expression through regulation of the intensity or duration of blue light exposure. In vitro studies showed that light-induced DTA expression reduced 4T1 cell viability and induced apoptosis. Furthermore, the LightOn gene expression system enabled spatiotemporal control of the expression of DTA in a mouse 4T1 tumor xenograft model, which resulted in excellent antitumor effects, reduced tumor angiogenesis, and no systemic toxicity. The combination of the LightOn gene expression system and NDDS may be an effective strategy for treatment of breast cancer.

11.
Br J Cancer ; 122(9): 1342-1353, 2020 04.
Article in English | MEDLINE | ID: mdl-32203206

ABSTRACT

BACKGROUND: Recent studies have shown that multidrug resistance may be induced by the high stemness of cancer cells. Following prolonged chemotherapy, MDR protein 1 (MDR1) and CD133 increase in CRC, but the relationship between them is unclear. METHODS: The relationship between MDR and CSC properties in CRC was determined via CCK-8 assay, apoptosis assay, DOX uptake and retention, immunohistochemistry, immunofluorescence and flow cytometry. The correlations between their expression levels were evaluated using Spearman's rank statistical test and the Mann-Whitney test. Furthermore, the effect of CD133 on the repression of the AKT/NF-κB/MDR1 signalling pathway was investigated in vitro and in vivo. RESULTS: We found that CD133 increased with the emergence of drug-resistance phenotypes, and the high expression of MDR1/P-gp was consistently accompanied by positive expression of CD133 as demonstrated by the analysis of patient samples. Up- or downregulation of CD133 could regulate MDR via AKT/NF-κB/MDR1 signalling in CRC. A rescue experiment showed that the AKT/NF-κB signalling pathway is the main mechanism by which CD133 regulates MDR1/P-gp expression in CRC. CONCLUSIONS: Taken together, our results suggest that targeting CD133 reverses drug resistance via the AKT/NF-κB/MDR1 pathway and that this pathway might serve as a potential therapeutic target to reverse MDR in CRC.


Subject(s)
AC133 Antigen/genetics , Colorectal Neoplasms/drug therapy , Doxorubicin/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , NF-kappa B/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Transcription Factor RelA/genetics
12.
Cancer Sci ; 111(5): 1619-1630, 2020 May.
Article in English | MEDLINE | ID: mdl-32058643

ABSTRACT

Recent studies have shown that MDR could be induced by the high stemness of cancer cells. In a previous study, we found bufalin could reverse MDR and inhibit cancer cell stemness in colorectal cancer, but the relationship between them was unclear. Here we identified overexpressing CD133 increases levels of Akt/nuclear factor-κB signaling mediators and MDR1, while increasing cell chemoresistance. Furthermore, bufalin reverses colorectal cancer MDR by regulating cancer cell stemness through the CD133/nuclear factor-κB/MDR1 pathway in vitro and in vivo. Taken together, our results suggest that bufalin could be developed as a novel 2-pronged drug that targets CD133 and MDR1 to eradicate MDR cells and could ultimately be combined with conventional chemotherapeutic agents to improve treatment outcomes for patients with colorectal cancer.


Subject(s)
AC133 Antigen/metabolism , Antineoplastic Agents/pharmacology , Bufanolides/pharmacology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Transcription Factor RelA/metabolism , AC133 Antigen/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antineoplastic Agents/therapeutic use , Bufanolides/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Multiple/drug effects , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Transcription Factor RelA/genetics
13.
Mol Pharm ; 17(1): 301-315, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31765570

ABSTRACT

The controversy surrounding the use of diphtheria toxin (DT) as a therapeutic agent against tumor cells arises mainly from its unexpected harmfulness to healthy tissues. We encoded the cytotoxic fragment A of DT (DTA) as an objective gene in the Light-On gene-expression system to construct plasmids pGAVPO (pG) and pU5-DTA (pDTA). Meanwhile, a cRGD-modified ternary complex comprising plasmids, chitosan, and liposome (pG&pDTA@cRGD-CL) was prepared as a nanocarrier to ensure transfection efficiency. Benefiting from spatiotemporal control of this light-switchable transgene system and the superior tumor targeting of the carrier, toxins were designed to be expressed selectively in illuminated lesions. In vitro studies suggested that pG&pDTA@cRGD-CL exerted arrest of the S phase in B16F10 cells upon blue light irradiation and, ultimately, induced the apoptosis and necrosis of tumor cells. Such DTA-based treatment exerted enhanced antitumor activity in mice bearing B16F10 xenografts and displayed prolonged survival time with minimal side effects. Hence, we described novel DTA-based therapy combined with nanotechnology and the Light-On gene-expression system: such treatment could be a promising strategy against melanoma.


Subject(s)
Diphtheria Toxin/genetics , Gene Expression/radiation effects , Genetic Therapy , Liposomes/chemistry , Melanoma, Experimental/therapy , Nanotechnology/methods , Peptide Fragments/genetics , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Cell Line, Tumor , Chitosan/chemistry , Gene Expression/genetics , Liposomes/ultrastructure , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Microscopy, Electron, Transmission , Particle Size , Peptides, Cyclic/chemistry , S Phase Cell Cycle Checkpoints/drug effects , S Phase Cell Cycle Checkpoints/genetics , S Phase Cell Cycle Checkpoints/radiation effects , Spheroids, Cellular/radiation effects , Tissue Distribution , Xenograft Model Antitumor Assays
14.
Int J Nanomedicine ; 14: 9631-9645, 2019.
Article in English | MEDLINE | ID: mdl-31824156

ABSTRACT

BACKGROUND: Photothermal therapy (PTT) has great potential in the clinical treatment of tumors. However, most photothermal materials are difficult to apply due to their insufficient photothermal conversion efficiencies (PCEs), poor photostabilities and short circulation times. Furthermore, tumor recurrence is likely to occur using PTT only. In the present study, we prepared cyclo (Arg-Gly-Asp-d-Phe-Cys) [c(RGD)] conjugated doxorubicin (DOX)-loaded Fe3O4@polydopamine (PDA) nanoparticles to develop a multifunctional-targeted nanocomplex for integrated tumor diagnosis and treatment. MATERIALS AND METHODS: Cytotoxicity of Fe3O4@PDA-PEG-cRGD-DOX against HCT-116 cells was determined by cck-8 assay. Cellular uptake was measured by confocal laser scanning microscope (CLSM). Pharmacokinetic performance of DOX was evaluated to compare the differences between free DOX and DOX in nanocarrier. Performance in magnetic resonance imaging (MRI) and antitumor activity of complex nanoparticles were evaluated in tumor-bearing nude mice. RESULTS: Fe3O4@PDA-PEG-cRGD-DOX has a particle size of 200-300 nm and a zeta potential of 22.7 mV. Further studies in vitro and in vivo demonstrated their excellent capacity to target tumor cells and promote drug internalization, and significantly higher cytotoxicity with respect to that seen in a control group was shown for the nanoparticles. In addition, they have good thermal stability, photothermal conversion efficiencies (PCEs) and pH responsiveness, releasing more DOX in a mildly acidic environment, which is very conducive to their chemotherapeutic effectiveness in the tumor microenvironment. Fe3O4@PDA-PEG-cRGD-DOX NPs were used in a subcutaneous xenograft tumor model of nude mouse HCT-116 cells showed clear signal contrast in T2-weighted images and effective anti-tumor chemo-photothermal therapy under NIR irradiation. CONCLUSION: According to our results, Fe3O4@PDA-PEG-cRGD-DOX had a satisfactory antitumor effect on colon cancer in nude mice and could be further developed as a potential integrated platform for the diagnosis and treatment of cancer to improve its antitumor activity against colon cancer.


Subject(s)
Doxorubicin/pharmacology , Ferric Compounds/chemistry , Hyperthermia, Induced , Indoles/chemistry , Magnetic Resonance Imaging , Nanocomposites/chemistry , Peptides, Cyclic/chemistry , Phototherapy , Polymers/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Combined Modality Therapy , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Drug Liberation , Endocytosis/drug effects , HCT116 Cells , Humans , Male , Mice, Nude , Nanocomposites/ultrastructure , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Neoplasm Recurrence, Local/drug therapy , Neoplasms/drug therapy , Neoplasms/pathology , Particle Size
15.
Mol Ther ; 27(10): 1810-1824, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31208913

ABSTRACT

Hypoxia is associated with poor prognosis and therapeutic resistance in cancer patients. Accumulating evidence has shown that microRNA (miRNA) plays an important role in the acquired drug resistance in colorectal carcinoma (CRC). However, the role of miRNA in hypoxia-induced CRC drug resistance remains to be elucidated. Here, we identified a hypoxia-triggered feedback loop that involves hypoxia-inducible transcription factor 1α (HIF-1α)-mediated repression of miR-338-5p and confers drug resistance in CRC. In this study, the unbiased miRNA array screening revealed that miR-338-5p is downregulated in both hypoxic CRC cell lines tested. Repression of miR-338-5p was required for hypoxia-induced CRC drug resistance. Furthermore, we identified interleukin-6 (IL-6), which mediates STAT3/Bcl2 activation under hypoxic conditions, as a direct miR-338-5p target. The resulting HIF-1α/miR-338-5p/IL-6 feedback loop was necessary for drug resistance in colon cancer cell lines. Using CRC patient samples, we found miR-338-5p has a negative correlation with HIF-1α and IL-6. Finally, in a xenograft model, overexpressing miR-338-5p in CRC cells and HIF-1α inhibitor PX-478 were able to enhance the sensitivity of CRC to oxaliplatin (OXA) via suppressing the HIF-1α/miR-338-5p/IL-6 feedback loop in vivo. Taken together, our results uncovered an HIF-1α/miR-338-5p/IL-6 feedback circuit that is critical in hypoxia-mediated drug resistance in CRC; targeting each member of this feedback loop could potentially reverse hypoxia-induced drug resistance in CRC.


Subject(s)
Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-6/genetics , MicroRNAs/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Colorectal Neoplasms/genetics , Feedback, Physiological , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mice , Neoplasm Transplantation , Oxaliplatin , Prognosis , Tumor Hypoxia
16.
J Biomed Nanotechnol ; 15(2): 329-339, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30596555

ABSTRACT

Bufalin has significant antitumor effects on various kinds of tumors. However, bufalin's clinical utility is severely limited because of its side effects, toxicity, and fast metabolism. We made bufalin-loaded, targeted nanospheres with the aim of overcoming bufalin's limitations. Epidermal growth factor (EGF)-modified nanospheres with encapsulated bufalin showed increased toxicity in colorectal cancer cells, inhibition of cell proliferation, and induction of apoptosis relative to the non-EGF-modified nanospheres. The average particle size of the nanospheres was 171 nm and the encapsulation efficiency was 83.2%. In vitro release data showed that bufalin loaded in calcium phosphate/DPPE-PEG-EGF hybrid porous nanospheres was released more slowly from dialysis membranes than free bufalin. According to our results, EGF-modified nanospheres containing bufalin show improved anti-tumor effects on colon cancer in nude mice, but without severe side effects.


Subject(s)
Nanospheres , Animals , Antineoplastic Agents , Bufanolides , Cell Line, Tumor , Epidermal Growth Factor , Mice , Mice, Nude , Phosphatidylethanolamines , Polyethylene Glycols
17.
Int J Nanomedicine ; 13: 7533-7548, 2018.
Article in English | MEDLINE | ID: mdl-30532537

ABSTRACT

BACKGROUND: Multidrug resistance (MDR) is the major reason for the failure of chemotherapy in colon cancer. Bufalin (BU) is one of the most effective antitumor active constituents in Chansu. Our previous study found that BU can effectively reverse P-glycoprotein (P-gp)-mediated MDR in colon cancer. However, the clinical application of BU is limited due to its low solubility in water and high toxicity. In the present study, a multifunctional delivery system based on vitamin-E- succinate grafted chitosan oligosaccharide (VES-CSO) and cyclic (arginine-glycine-aspartic acid peptide) (RGD)-modified d-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS) was prepared by emulsion solvent evaporation method for targeted delivery of BU to improve the efficacy of drug-resistant colon cancer therapy. METHODS: The cytotoxicity of BU-loaded micelles against drug-resistant colon cancer LoVo/ADR and HCT116/LOHP cells was measured by CCK-8 assay. The cellular uptake, Rho123 accumulation, and cell apoptosis were determined by flow cytometry. The expression of apoptosis-related protein and P-gp was measured by Western blot assay. The antitumor activity of BU-loaded micelles was evaluated in LoVo/ADR-bearing nude mice. RESULTS: BU-loaded VES-CSO/TPGS-RGD mixed micelles (BU@VeC/T-RGD MM) were 140.3 nm in diameter with zeta potential of 8.66 mV. The BU@VeC/T-RGD MM exhibited good stability, sustained-release pattern, higher intracellular uptake, and greater cytotoxicity in LoVo/ADR cells. Furthermore, the mechanisms of the BU@VeC/T-RGD MM to overcome MDR might be due to enhanced apoptosis rate and P-gp efflux inhibition. Subsequently, in vivo studies confirmed an enhanced therapeutic efficiency and reduced side effects associated with BU@VeC/T-RGD MM compared with free BU, owing to the enhanced permeation and retention effect, improved pharmacokinetic behavior, and tumor targeting, which lead to MDR-inhibiting effect in LoVo/ADR-bearing nude mice. CONCLUSION: Our results demonstrated that VeC/T-RGD MM could be developed as a potential delivery system for BU to improve its antitumor activity against drug-resistant colon cancer.


Subject(s)
Bufanolides/therapeutic use , Chitosan/chemistry , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Micelles , Oligopeptides/chemistry , Oligosaccharides/chemistry , Vitamin E/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bufanolides/pharmacology , Colonic Neoplasms/pathology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , HCT116 Cells , Humans , Mice, Nude , Rhodamine 123/metabolism , Tumor Burden
18.
Oncotarget ; 8(29): 48012-48026, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28624793

ABSTRACT

Multidrug resistance (MDR), mainly mediated by ABCB1 transporter, is a major cause for chemotherapy failure. Bufalin (BU), an active component of the traditional Chinese medicine chan'su, has been reported to have antitumor effects on various types of cancer cells. The purpose of this present study was to investigate the reversal effect of BU on ABCB1-mediated multidrug resistance in colorectal cancer. BU at safe concentration (5, 10, 20 nM) could reverse chemosensitivity of ABCB1-overexpression HCT8/ADR, LoVo/ADR and HCT8/ABCB1 nearly back to their parental cells level. In addition, results from the drug accumulation studies revealed that BU was able to enhance intracellular accumulation of doxorubicin (DOX) and Rhodamine 123 (Rho-123) in a dose-dependent manner. Furthermore, Western blot assays showed that BU significantly inhibited the expression level of ABCB1 protein. Meanwhile, BU stimulated the ATPase activity of ABCB1, which suggested that BU might be a substrate of ABCB1. More interestingly, docking analysis predicted that BU could be docked into the large hydrophobic drug-binding cavity of human ABCB1. Importantly, BU remarkable increased the effect of DOX against the ABCB1 resistant HCT8/ADR colorectal cell xenografts in nude mice, without inducing any obvious toxicity. Overall, we concluded that BU efficiently reversed ABCB1-mediated MDR through not only inhibited the efflux function of ABCB1, but also down-regulate its protein expression, which might represent a potential and superior ABCB1 modulator in colorectal cancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents/pharmacology , Bufanolides/pharmacology , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Animals , Antineoplastic Agents/chemistry , Bufanolides/chemistry , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Mice , Models, Molecular , Molecular Conformation , Protein Binding , Xenograft Model Antitumor Assays
19.
Oncotarget ; 8(13): 21719-21732, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423513

ABSTRACT

The development of multidrug-resistance (MDR) is a major contributor to death in colorectal carcinoma (CRC). Here, we investigated the possible role of microRNA (miR)-503-5p in drug resistant CRC cells. Unbiased microRNA array screening revealed that miR-503-5p is up-regulated in two oxaliplatin (OXA)-resistant CRC cell lines. Overexpression of miR-503-5p conferred resistance to OXA-induced apoptosis and inhibition of tumor growth in vitro and in vivo through down-regulation of PUMA expression. miR-503-5p knockdown sensitized chemoresistant CRC cells to OXA. Our studies indicated that p53 suppresses miR-503-5p expression and that deletion of p53 upregulates miR-503-5p expression. Inhibition of miR-503-5p in p53 null cells increased their sensitivity to OXA treatment. Importantly, analysis of patient samples showed that expression of miR-503-5p negatively correlates with PUMA in CRC. These results indicate that a p53/miR-503-5p/PUMA signaling axis regulates the CRC response to chemotherapy, and suggest that miR-503-5p plays an important role in the development of MDR in CRC by modulating PUMA expression.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins/metabolism , Adult , Aged , Animals , Blotting, Western , Drug Resistance, Multiple/genetics , Female , HCT116 Cells , HT29 Cells , Heterografts , Humans , Male , Mice , Mice, Nude , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
20.
Oncol Rep ; 37(3): 1815-1825, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28184922

ABSTRACT

Cinobufagin (CBF) is isolated from the skin and posterior auricular glands of the Asiatic toad (Bufo gargarizans). This study investigated the reversal effect of CBF on P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) in colon cancer. The effect of CBF on the cytotoxicity of anticancer drugs in P-gp overexpressing LoVo/ADR, HCT116/L, Cao-2/ADR cells and their parental cells was determined using CCK-8 assay. Apoptosis of anti-cancer drugs and accumulation of doxorubicin (DOX) and Rhodamine 123 (Rho123) in P-gp overexpressing cells were evaluated by flow cytometry. Results indicated that CBF significantly enhanced the sensitivity of P-gp substrate drugs on P-gp overexpressing cells, but had no effect on their parental cells. CBF enhanced the effect of DOX against P-gp-overexpressing LoVo/ADR cell xenografts in nude mice. Moreover, CBF also increased cell apoptosis of chemotherapy agents and intracellular accumulation of DOX and Rho123 in the MDR cells. Further research on the mechanisms revealed non-competitive inhibition of P-gp ATPase activity, but without altering the expression of P-gp. These findings demonstrated that CBF could be further developed into a safe and potent P-gp modulator for combination use with anticancer drugs in cancer chemotherapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Bufanolides/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Doxorubicin/pharmacology , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...