Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Pharm ; 606: 120872, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34246743

ABSTRACT

Doxorubicin (DOX) has been widely incorporated in various delivery forms for tareted treatment of malignant tumors such as triple-negative breast cancer (TNBC), with numerous studies reporting higher therapeutic efficacy and lower toxicity at the same time. However, little attention has been paid to whether DOX in a delivery form acts with the same actions and processes as in free form at the cellular level. This question was investigated in the present study wherein DOX conjugated with polyglycerol-coated nanodiamonds through the pH-sensitive hydrazone bond (Nano-DOX) was compared with DOX in free form on the 4T1 mouse TNBC model. We first found Nano-DOX to have a distinct intracellular distribution profile from DOX. Internalized Nano-DOX mainly stayed in the lysosomes slowly releasing DOX into the cytoplasm and then the nucleus whereas DOX displayed both nuclear and lysosomal distribution after cell uptake. Next, Nano-DOX was shown to induce endoplasmic reticulum (ER) stress without substantial DNA damage while DOX caused massive DNA damage as well as ER stress. Consequently, Nano-DOX only caused minimal activation of pro-inflammatory signaling mediated by MAPK/ERK, NF-κB and STAT3 as seen in response to DOX-inflicted DNA damage. Consistently, DOX-induced activities of ABC transporters, CXCL-1, GM-CSF and IL-6, which are tumor protective events downstream to the pro-inflammatory signaling, were also minimal in Nano-DOX-treated cancer cells. These findings are compelling proof that a chemotherapy in nano form can have distinct intracellular pharmacokinetics from its free from, which can result in altered cellular effects of the drug. Implications of these findings are discussed with an emphasis on nano-drug design, tumor pharmacology and chemoresistance.


Subject(s)
Nanodiamonds , Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Doxorubicin , Humans , Hydrazones , Mice
2.
Biomed Pharmacother ; 132: 110867, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33075668

ABSTRACT

Hepatocellular Carcinoma (HCC) is one of the most common malignant tumors, and high recurrence and metastasis are the major obstacles to successful treatment of HCC. Traditional Chinese medicine has little known and unique advantages in the treatment of HCC. Previous studies have confirmed that Chinese herbal formula Qingrejiedu (clears away heat and toxins), Huoxuehuayu (promotes blood flow to remove stasis) and Fuzhengguben (strengthens healthy qi and root) (QHF) has a significant effect on patients with advanced HCC, improves the quality of life and prolongs the survival time of patients significantly. In this study, we investigated the effect of QHF on proliferation, migration and invasion of human high metastatic hepatocellular carcinoma cell line HCCLM3 and its underlying mechanism. The results from our in vitro experiments showed that QHF has the ability to inhibit the proliferation by inducing G2/M phase cell cycle arrest and induce apoptosis. Moreover, QHF can also inhibit migration and invasion of HCCLM3 cells and the expression of the p-c-Met protein in HCCLM3 cells was down-regulated. c-Met is closely related to the metastasis of HCC, then we constructed a stable transfected cell line HepG2-met with high expression of c-Met by transfection. Further study in vivo revealed that c-Met gene will promote the growth of tumors and lung metastases in nude mice, and QHF intervention can reduce tumor lung metastases by inhibiting the HGF/c-Met signaling pathway. In conclusion, our study reveals that QHF can inhibit the proliferation, migration and invasion of HCCLM3, and this effect may be related to inhibiting HGF/c-Met signaling pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drugs, Chinese Herbal/pharmacology , Hepatocyte Growth Factor/metabolism , Liver Neoplasms/drug therapy , Lung Neoplasms/prevention & control , Proto-Oncogene Proteins c-met/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Cell Movement/drug effects , Cell Proliferation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Hepatocyte Growth Factor/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Proto-Oncogene Proteins c-met/genetics , Signal Transduction , Xenograft Model Antitumor Assays
3.
J Control Release ; 320: 469-483, 2020 04 10.
Article in English | MEDLINE | ID: mdl-31987922

ABSTRACT

Astrocytes are key stromal components in glioblastoma (GBM) and have complex interactions with the GBM cells (GBC) promoting the survival, progression and therapy resistance of GBM. In this study, we first demonstrated the existence of a reciprocal activation loop mediated by the STAT3/IL-6 signaling between GBC and astrocytes. This loop of reciprocity was found to be initiated by the constitutive activity of STAT3 and downstream expression of IL-6 in the GBC. GBC-derived IL-6 activated STAT3 and thereby upregulated IL-6 expression in the astrocytes. Astrocyte-derived IL-6 acted back on the GBC causing further activation of STAT3 and leading to enhanced downstream events that promote proliferation, migration, invasion and apoptosis resistance of the GBC. Next, we showed that doxorubicin-polyglycerol-nanodiamond conjugates (Nano-DOX), which could be delivered via GBM-associated macrophages, suppressed STAT3 activity in the GBC reducing their IL-6 output to the astrocytes and thereby abolished the astrocytes' feedback activation of the GBC. Moreover, Nano-DOX also suppressed stimulated activation of STAT3 and IL-6 induced by temozolomide, a first-line anti-GBM chemotherapy, resistance to which critically involves STAT3 activation. In conclusion, Nano-DOX could disrupt the STAT3/IL-6-mediated reciprocal activation loop between the GBC and astrocytes. Nano-DOX also provides a novel approach to therapeutic modulation of the GBM microenvironment.


Subject(s)
Glioblastoma , Nanodiamonds , Astrocytes/metabolism , Cell Line, Tumor , Cell Proliferation , Doxorubicin/pharmacology , Glioblastoma/drug therapy , Glycerol , Humans , Interleukin-6 , Polymers , STAT3 Transcription Factor/metabolism , Tumor Microenvironment
4.
J Nanobiotechnology ; 17(1): 110, 2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31623629

ABSTRACT

BACKGROUND: Triple negative breast cancer (TNBC) has the poorest prognosis of all breast cancer subtypes and is one of the most fatal diseases for women. Combining cytotoxic chemotherapy with immunotherapy has shown great promise for TNBC treatment. However, chemotherapy often leads to the development of chemoresistance and severe systemic toxicity compromising the immune functions that are crucial to anti-TNBC immune therapy. Tumor-induced immunosuppression also poses a great hindrance to efficacious anti-TNBC immunotherapy. Nanomedicine holds great promise to overcome these hurdles. RESULTS: Doxorubicin-polyglycerol-nanodiamond conjugate (Nano-DOX) was firstly found to be a cytostatic agent to the 4T1 cells and displayed a lower apparent therapeutic potency than DOX. However, the tumor-bearing animals, particularly some key immune cells thereof, showed good tolerance of Nano-DOX as opposed to the severe toxicity of DOX. Next, Nano-DOX did not induce significant upregulation of P-gp and IL-6, which were demonstrated to be key mediators of chemoresistance to DOX in the 4T1 cells. Then, Nano-DOX was shown to downregulate tumor-derived granulocyte-colony stimulating factor (G-CSF) and suppresses the induction and tissue filtration of myeloid-derived suppressor cells (MDSCs) that are the principal effectors of cancer-associated systemic immunosuppression. Nano-DOX also alleviated the phenotype of MDSCs induced by 4T1 cells. Finally, Nano-DOX induced the 4T1 cells to emit damage associated molecular patterns (DAMPs) that stimulated the tumor immune microenvironment through activating key immune effector cells involved in anti-tumor immunity, such as macrophages, dendritic cells and lymphocytes in the tumor tissue. CONCLUSIONS: Nano-DOX is a cytostatic agent with good host tolerance which is capable of evading chemoresistance and reversing cancer-induced immunosuppression both at the systemic level and in the tumor microenvironment in TNBC. Our work presents Nano-DOX as an interesting example that a chemotherapeutic agent in nano-form may possess distinct biochemical properties from its free form, which can be exploited to join chemotherapy with immunotherapy for better treatment of cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cytostatic Agents/administration & dosage , Doxorubicin/administration & dosage , Glycerol/chemistry , Nanoconjugates/chemistry , Polymers/chemistry , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cytostatic Agents/therapeutic use , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Humans , Immune Tolerance/drug effects , Mice, Inbred BALB C , Nanodiamonds/chemistry , Triple Negative Breast Neoplasms/immunology , Tumor Microenvironment/drug effects
5.
Acta Biomater ; 86: 381-394, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30654213

ABSTRACT

Immunosuppression is a salient feature of GBM associated with the disease's grim prognosis and the limited success of anti-GBM immunotherapy. Stimulating immunogenicity of the GBM cells (GC) is a promising approach to subverting the GBM-associated immunosuppression. We had previously devised a drug composite based on polyglycerol-functionalized nanodiamonds bearing doxorubicin (Nano-DOX) and demonstrated that Nano-DOX effectively modulated GBM's immunosuppressive microenvironment through stimulating the immunogenicity of GC and initiated anti-GBM immune responses. The present study now explored the mechanism of Nano-DOX's immunostimulatory action. Nano-DOX was found to induce autophagy rather than apoptosis in GC and stimulated GC to emit antigens and damage-associated molecular patterns (DAMPs) that are potent adjuvants, which resulted in enhanced activation of dendritic cells (DC). Heightened autophagosome release was observed in Nano-DOX-treated GC but was shown not to be a major channel of antigen donation. Blocking autophagy in GC not only reduced Nano-DOX-stimulated GC antigen donation and DAMPs emission, but also efficiently attenuated DC activation stimulated by Nano-DOX-treated GC. Taken together, these findings suggest that activation of autophagy is a central mechanism whereby Nano-DOX stimulates GC's immunogenicity. Our work provides new insight on how nanotechnology can be applied to therapeutically modulate the GBM immune microenvironment by harnessing autophagy in the cancer cells. STATEMENT OF SIGNIFICANCE: Immunosuppression is a salient feature of GBM associated with the grim prognosis of the disease and the limited success of anti-GBM immunotherapy. We demonstrated that Doxorubicin-polyglycerol-nanodiamond composites could activate autophagy in GBM cells and thereby stimulate the immunogenecity of GBM cells. This discovery 1, sheds new light on how nanotechnology could be applied to therapeutically modulate the tumor immune microenvironment, and 2, provides a powerful tool for subverting the GBM's immunosuppressive microenvironment, which has great therapeutic potential for the treatment of GBM.


Subject(s)
Autophagy , Doxorubicin/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/immunology , Glycerol/chemistry , Nanodiamonds/chemistry , Polymers/chemistry , Animals , Antigen Presentation/drug effects , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Doxorubicin/pharmacology , Female , Glioblastoma/pathology , Humans , Mice, Inbred BALB C , Mice, Nude , Nanodiamonds/ultrastructure , Pathogen-Associated Molecular Pattern Molecules/metabolism , THP-1 Cells
6.
Biomaterials ; 181: 35-52, 2018 10.
Article in English | MEDLINE | ID: mdl-30071380

ABSTRACT

Glioblastoma (GBM) is the deadliest and most common type of primary brain tumor in adults with a grim prognosis despite multimodal treatments. Dendritic cell (DC)-based immunotherapy has emerged as a promising therapeutic modality for GBM, whose efficacy is nonetheless fundamentally undermined by GBM-induced immunosuppression. Inducing emission of damage associated molecular patterns (DAMPs) is a highly effective strategy to subvert tumor-associated immunosuppression. The present work was carried out to explore the idea of subverting the GBM immunosuppressive microenvironment through DC-mediated delivery of doxorubicin-polyglycerol-nanodiamond composites (Nano-DOX), a potent DAMPs inducer demonstrated by our previous study, and thereby eliciting enhanced DC-driven anti-GBM immune response. In the in-vitro work on human cell models, Nano-DOX-loaded DC were shown to be functionally viable and release cargo drug to co-cultured GBM cells (GC). Nano-DOX-treated GC displayed not only profuse DAMPs emission but also antigen release. Enhanced activation and acquisition and presentation of GC-derived antigen were then demonstrated in DC in co-culture with GC and Nano-DOX. Consistently, co-culture with GC and Nano-DOX also activated mouse bone marrow-derived DC (mDC) which in turn stimulated mouse spleen-derived lymphocytes which ultimately suppressed co-cultured GC. Next, athymic mice bearing orthotopic human GBM xenografts were intravenously injected with Nano-DOX-loaded mDC and, 48 h later, spleen-derived lymphocytes. The presence of Nano-DOX, DAMPs emission and enhanced infiltration and activation of mDC and lymphocytes were detected in the GBM xenografts. Taken together, our results demonstrate the efficacy of DC-mediated delivery of Nano-DOX to stimulate GC immunogenicity and elicit anti-cancer immune response in the GBM. By this work, we present a novel approach with great application potential to subverting the GBM immunosuppressive microenvironment and to anti-GBM immunotherapy. Investigation has also been conducted probing the mechanisms by which Nano-DOX stimulates GC immunogenicity, which is described in a follow-up paper.


Subject(s)
Brain Neoplasms/drug therapy , Dendritic Cells/metabolism , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Glycerol/chemistry , Polymers/chemistry , Animals , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/adverse effects , Drug Carriers/chemistry , Female , Glioblastoma , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanostructures/adverse effects , Nanostructures/chemistry , THP-1 Cells
7.
Oncotarget ; 8(7): 11425-11441, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28002791

ABSTRACT

OBJECTIVES: To investigate the mechanism of cinobufagin-reduced cancer pain in mouse cancer pain model and in vitro cell co-culture system. METHODS: Female Kunming mice were randomly divided into 4 groups. One group of animals was set as normal control without any treatment. Other three groups of animals received H22 hepatoma cell inoculation in right hind paw. At day 9 after inoculation, mice in other three groups were injected intraperitoneally once a day for 8 days with the solvent, morphine or cinobufagin, respectively. The pain behavior was recorded daily. On the last day, all mice were sacrificed and xenograft tissues homogenate and plasma levels of ß-endorphin (ß-END), corticotropin-releasing factor (CRF) and interleukin-1ß (IL-1ß) were assessed by ELISA assay. Immunohistochemistry was performed to determine the expression of ß-END, pro-opiomelanocortin (POMC) and the µ-opioid receptor (µ-OR) in the xenograft tissues. Immunofluorescence was used to localize lymphocytes with expression of CD3+, CD4+ and CD8+ in xenograft tumors and adjacent tissues. Mice splenic lymphocytes and H22 hepatoma carcinoma ascites cells were prepared for co-culture. ß-END and CRF were detected in co-culture supernatants. The MTT assay and cytometry were used to assess cell proliferation. RT-PCR was conducted to determine the gene expression of POMC and Cathepsin L (CTSL). Chemotaxis was examined using a transwell-based migration assay. RESULTS: Compared to the model group, the thermal and mechanical pain thresholds were increased in mice after cinobufagin treatment. The expression of ß-END and CRF in the plasma and tumor tissues of cinobufagin group were much higher than that of the model group mice, but the expression of IL-1ß in the plasma and tumor tissues was much lower than that in the model group mice. Meanwhile, the expression of ß-END, POMC and µ-OR proteins was significantly increased in the xenograft tissues from cinobufagin group. Lymphocyte population of CD3+, CD4+, CD8+ were also elevated in xenograft tumors and adjacent tissues. In the cell co-culture assays, the content of ß-END in the supernatant was significantly increased by cinobufagin in a dose-dependent manner. Cinobufagin also largely increased the proliferation of immune cells and inhibited H22 hepatoma carcinoma cell proliferation in single or co-culture cell assays. Gene expression of POMC and CTSL in cinobufagin group was significantly up-regulated comparing to the control group. Finally, cinobufagin addition enhanced the migration of immune cells in transwell assay. CONCLUSIONS: Cinobufagin-induced local analgesic effect might be associated with increased activity of POMC/ß-END/µ-OR pathway released from invaded CD3/4/8 lymphocytes in cancer tissues.


Subject(s)
Analgesics/pharmacology , Bufanolides/pharmacology , Neoplasms, Experimental/complications , Pain/drug therapy , Pain/etiology , Animals , Carcinoma, Hepatocellular/complications , Cell Line, Tumor , Coculture Techniques , Corticotropin-Releasing Hormone/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Interleukin-1beta/metabolism , Liver Neoplasms/complications , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Pain Threshold , Polymerase Chain Reaction , Random Allocation , beta-Endorphin/metabolism
8.
Int Immunopharmacol ; 42: 90-99, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27907881

ABSTRACT

The changes in thermal and mechanical hyperalgesia in paw cancer pain model mice and the action mechanism of toad skin extracts (TSE) was investigated. Eighty female mice were subcutaneously injected with saline or inoculated with H22 hepatoma cells in the right hind paw and administration with saline, vehicle, morphine and TSE. The pain behavior was recorded before treatment and at 0.5, 1.0, 1.5, 3 and 6h after initial administration, and thereafter on the 2nd, 4th, 6th, and 8th day after administration. On the last day, samples were collected after the euthanasia for the detection of ß-END, CRF, IL-1ß, POMC, µ-OR, CD3+, CD8+ and CD4+ in sera and the tumor tissues. The results showed that TSE significantly increased the thresholds of thermal pain and mechanical pain, and upregulated the expressions of ß-END, CRF, POMC, CD3+, CD8+ and µ-OR, and downregulated the expression of CD4+. These results indicate that TSE significantly relieved pain in cancer pain model mice and raised their pain threshold. In addition, TSE seems to play a prominent role in promoting the activity of tumor infiltrating lymphocytes (TILs, CD3+ and CD8+ T cells), and this immune-cell-derived peripheral analgesic pathway might have widespread potential for clinical use.


Subject(s)
Anura/physiology , Cell Extracts/therapeutic use , Liver Neoplasms, Experimental/therapy , Liver Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Pain Management , Skin Neoplasms/therapy , Skin/cytology , Animals , CD8 Antigens/metabolism , Female , Humans , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/pathology , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred Strains , Pain Measurement , Pain Threshold/drug effects , Skin Neoplasms/pathology
9.
World J Gastroenterol ; 21(35): 10126-36, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26401077

ABSTRACT

AIM: To study the effects of QHF-cisplatin on H22 hepatocellular carcinoma (HCC) and their mechanisms of action. METHODS: Sixty BALB/c mice were randomly divided into a model group (n = 48) and a normal control group (n = 12). An HCC xenograft tumor was created by injecting H22 cells directly into the liver parenchyma of the mice. The 48 BALB/c mice in the model group were randomly divided into four groups: QHF, DDP (cisplatin), QHF plus DDP, and model control. The inhibitory effects of these drugs on tumor growth were evaluated by calculating the rate of tumor growth inhibition. The mice were examined by observing their general condition, body weight and survival time. Changes in tumor tissue were observed under an optical microscope. Aspartate aminotransferase (AST), alanine aminotransferase (ALT) and α-fetoprotein (AFP) levels in serum were measured. Hepatocyte growth factor (HGF), c-mesenchymal-epithelial transition (c-Met) factor, phosphorylated (p)-c-Met, p38, p-p38, extracellular signal-regulated kinase (ERK), p-ERK and vascular endothelial growth factor (VEGF) levels were evaluated in tumor and liver tissues using western blotting. RESULTS: Compared with the DDP group, a lower incidence of toxic reactions and a higher survival time were observed in the QHF plus DDP group. Tumor weight was significantly lower in the QHF, DDP and QHF plus DDP groups than in the model control group (0.24 ± 0.07, 0.18 ± 0.03 and 0.14 ± 0.01 g vs 0.38 ± 0.05 g, respectively), and the differences were statistically significant (P < 0.01). The rate of tumor growth inhibition in the QHF, DDP and QHF plus DDP groups was 38.7%, 52.6% and 63.5%, respectively. AST, ALT and AFP levels in serum were significantly lower in the QHF, DDP and QHF plus DDP groups compared to the model control group (P < 0.05). Similarly, HGF, p-c-Met, p-p38, p-ERK and VEGF levels in tumor tissue were significantly lower in the QHF, DDP and QHF plus DDP groups (P < 0.05). CONCLUSION: QHF and DDP have an antiangiogenic effect on H22 HCC in mice. QHF inhibits tumor growth via blocking the HGF/c-Met signaling pathway, inhibiting p38, ERK and VEGF signaling.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cisplatin/pharmacology , Drugs, Chinese Herbal/pharmacology , Liver Neoplasms, Experimental/drug therapy , Animals , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Female , Liver Neoplasms, Experimental/blood , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/pathology , Male , Mice, Inbred BALB C , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...