Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.993
Filter
1.
Clin Exp Pharmacol Physiol ; 51(7): e13900, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38843865

ABSTRACT

Traditional Chinese medicine, specifically the Jianpi Tiaoqi (JPTQ) decoction, has been explored for its role in treating breast cancer, particularly in inhibiting lung metastasis in affected mice. Our study evaluated the effects of JPTQ on several factors, including tumour growth, apoptosis, angiogenesis, epithelial-to-mesenchymal transition (EMT) and immune microenvironment regulation. We used bioluminescence imaging to observe in situ tumour growth and potential lung metastasis. Transcriptomic analysis provided insights into gene expression, whereas flow cytometry was used to examine changes in specific immune cells, such as CD4+ T cells and myeloid-derived suppressor cells. Several essential proteins and genes, including vascular endothelial growth factor (VEGF), matrix metalloprotein-9 (MMP-9) and B-cell lymphoma 2 (Bcl-2), were assessed through quantitative real-time polymerase chain reaction, western blotting and immunohistochemistry. Our findings showed that JPTQ treatment inhibited tumour proliferation in cancer-bearing mice. Bioluminescence imaging and pathological analysis indicated a reduction in lung metastasis. Transcriptome analysis of lung and tumour tissues indicated that the genes associated with EMT, angiogenesis, proliferation and apoptosis were regulated in the JPTQ-treated group. Kyoto Encyclopedia of Genes and Genomes analysis suggested enrichment of immune-related pathways. Flow cytometry indicated that JPTQ treatment reduced the proportion of monocyte-myeloid-derived suppressor cells in the lung and increased the number of CD4+ T cells in the peripheral blood and the number of T helper 1 (Th1) cells in the spleen (P < 0.05). E-cadherin and cleaved caspase 3 were upregulated, whereas Snail, Bcl-2, Ki67 and VEGF were downregulated in the lung and tumour tissues; moreover, the expression of MMP-9 was downregulated in the lung tissue (P < 0.05). In essence, JPTQ not only inhibits tumour growth in affected mice, but also promotes positive immune responses, reduces angiogenesis, boosts tumour cell apoptosis, reverses EMT and decreases breast cancer lung metastasis.


Subject(s)
Cell Proliferation , Drugs, Chinese Herbal , Epithelial-Mesenchymal Transition , Lung Neoplasms , Triple Negative Breast Neoplasms , Animals , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Lung Neoplasms/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Mice , Cell Proliferation/drug effects , Female , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Cell Line, Tumor , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Tumor Microenvironment/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
2.
PLoS One ; 19(6): e0304782, 2024.
Article in English | MEDLINE | ID: mdl-38833447

ABSTRACT

PURPOSE: Pathological angiogenesis and vascular instability are observed in diabetic retinopathy (DR), diabetic macular edema (DME), and wet age-related macular degeneration (wAMD). Many receptor tyrosine kinases (RTKs) including vascular endothelial growth factor receptors (VEGFRs) contribute to angiogenesis, whereas the RTK TIE2 is important for vascular stability. Pan-VEGFR tyrosine kinase inhibitors (TKIs) such as vorolanib, sunitinib, and axitinib are of therapeutic interest over current antibody treatments that target only one or two ligands. This study compared the anti-angiogenic potential of these TKIs. METHODS: A kinase HotSpot™ assay was conducted to identify TKIs inhibiting RTKs associated with angiogenesis and vascular stability. Half-maximal inhibitory concentration (IC50) for VEGFRs and TIE2 was determined for each TKI. In vitro angiogenesis inhibition was investigated using a human umbilical vein endothelial cell sprouting assay, and in vivo angiogenesis was studied using the chorioallantoic membrane assay. Melanin binding was assessed using a melanin-binding assay. Computer modeling was conducted to understand the TIE2-axitinib complex as well as interactions between vorolanib and VEGFRs. RESULTS: Vorolanib, sunitinib, and axitinib inhibited RTKs of interest in angiogenesis and exhibited pan-VEGFR inhibition. HotSpot™ assay and TIE2 IC50 values showed that only axitinib potently inhibited TIE2 (up to 89%). All three TKIs effectively inhibited angiogenesis in vitro. In vivo, TKIs were more effective at inhibiting VEGF-induced angiogenesis than the anti-VEGF antibody bevacizumab. Of the three TKIs, only sunitinib bound melanin. TKIs differ in their classification and binding to VEGFRs, which is important because type II inhibitors have greater selectivity than type I TKIs. CONCLUSIONS: Vorolanib, sunitinib, and axitinib exhibited pan-VEGFR inhibition and inhibited RTKs associated with pathological angiogenesis. Of the three TKIs, only axitinib potently inhibited TIE2 which is an undesired trait as TIE2 is essential for vascular stability. The findings support the use of vorolanib for therapeutic inhibition of angiogenesis observed in DR, DME, and wAMD.


Subject(s)
Angiogenesis Inhibitors , Axitinib , Human Umbilical Vein Endothelial Cells , Imidazoles , Indazoles , Indoles , Protein Kinase Inhibitors , Pyrroles , Receptors, Vascular Endothelial Growth Factor , Sunitinib , Axitinib/pharmacology , Humans , Sunitinib/pharmacology , Angiogenesis Inhibitors/pharmacology , Imidazoles/pharmacology , Pyrroles/pharmacology , Indoles/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Indazoles/pharmacology , Animals , Protein Kinase Inhibitors/pharmacology , Receptor, TIE-2/metabolism , Receptor, TIE-2/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism
3.
Int J Biol Sci ; 20(7): 2454-2475, 2024.
Article in English | MEDLINE | ID: mdl-38725854

ABSTRACT

The emergence of Poly (ADP-ribose) polymerase inhibitors (PARPi) has marked the beginning of a precise targeted therapy era for ovarian cancer. However, an increasing number of patients are experiencing primary or acquired resistance to PARPi, severely limiting its clinical application. Deciphering the underlying mechanisms of PARPi resistance and discovering new therapeutic targets is an urgent and critical issue to address. In this study, we observed a close correlation between glycolysis, tumor angiogenesis, and PARPi resistance in ovarian cancer. Furthermore, we discovered that the natural compound Paris saponin VII (PS VII) partially reversed PARPi resistance in ovarian cancer and demonstrated synergistic therapeutic effects when combined with PARPi. Additionally, we found that PS VII potentially hindered glycolysis and angiogenesis in PARPi-resistant ovarian cancer cells by binding and stabilizing the expression of RORα, thus further inhibiting ECM1 and interfering with the VEGFR2/FAK/AKT/GSK3ß signaling pathway. Our research provides new targeted treatment for clinical ovarian cancer therapy and brings new hope to patients with PARPi-resistant ovarian cancer, effectively expanding the application of PARPi in clinical treatment.


Subject(s)
Diosgenin/analogs & derivatives , Glycolysis , Neovascularization, Pathologic , Ovarian Neoplasms , Saponins , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2 , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Saponins/pharmacology , Saponins/therapeutic use , Signal Transduction/drug effects , Glycolysis/drug effects , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Drug Resistance, Neoplasm/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Animals , Mice, Nude , Mice , Angiogenesis
4.
Mol Med ; 30(1): 57, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698308

ABSTRACT

BACKGROUND: Ossification of the posterior longitudinal ligament (OPLL), an emerging heterotopic ossification disease, causes spinal cord compression, resulting in motor and sensory dysfunction. The etiology of OPLL remains unclear but may involve integrin αVß3 regulating the process of osteogenesis and angiogenesis. In this study, we focused on the role of integrin αVß3 in OPLL and explored the underlying mechanism by which the c(RGDyk) peptide acts as a potent and selective integrin αVß3 inhibitor to inhibit osteogenesis and angiogenesis in OPLL. METHODS: OPLL or control ligament samples were collected in surgery. For OPLL samples, RNA-sequencing results revealed activation of the integrin family, particularly integrin αVß3. Integrin αVß3 expression was detected by qPCR, Western blotting, and immunohistochemical analysis. Fluorescence microscopy was used to observe the targeted inhibition of integrin αVß3 by the c(RGDyk) peptide on ligaments fibroblasts (LFs) derived from patients with OPLL and endothelial cells (ECs). The effect of c(RGDyk) peptide on the ossification of pathogenic LFs was detected using qPCR, Western blotting. Alkaline phosphatase staining or alizarin red staining were used to test the osteogenic capability. The effect of the c(RGDyk) peptide on angiogenesis was determined by EC migration and tube formation assays. The effects of the c(RGDyk) peptide on heterotopic bone formation were evaluated by micro-CT, histological, immunohistochemical, and immunofluorescence analysis in vivo. RESULTS: The results indicated that after being treated with c(RGDyk), the osteogenic differentiation of LFs was significantly decreased. Moreover, the c(RGDyk) peptide inhibited the migration of ECs and thus prevented the nutritional support required for osteogenesis. Furthermore, the c(RGDyk) peptide inhibited ectopic bone formation in mice. Mechanistic analysis revealed that c(RGDyk) peptide could inhibit osteogenesis and angiogenesis in OPLL by targeting integrin αVß3 and regulating the FAK/ERK pathway. CONCLUSIONS: Therefore, the integrin αVß3 appears to be an emerging therapeutic target for OPLL, and the c(RGDyk) peptide has dual inhibitory effects that may be valuable for the new therapeutic strategy of OPLL.


Subject(s)
Integrin alphaVbeta3 , Ossification of Posterior Longitudinal Ligament , Osteogenesis , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Humans , Osteogenesis/drug effects , Animals , Mice , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification of Posterior Longitudinal Ligament/drug therapy , Male , Female , Middle Aged , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Neovascularization, Physiologic/drug effects , Cell Movement/drug effects , Disease Models, Animal , Oligopeptides/pharmacology , Oligopeptides/chemistry , Angiogenesis
5.
Drug Dev Res ; 85(3): e22195, 2024 May.
Article in English | MEDLINE | ID: mdl-38704831

ABSTRACT

We investigated the angiogenesis-modulating ability of noscapine in vitro using osteosarcoma cell line (MG-63) and in vivo using a zebrafish model. MTT assay and the scratch wound healing assay were performed on the osteosarcoma cell line (MG-63) to analyze the cytotoxic effect and antimigrative ability of noscapine, respectively. We also observed the antiangiogenic ability of noscapine on zebrafish embryos by analyzing the blood vessels namely the dorsal aorta, and intersegmental vessels development at 24, 48, and 72 h postfertilization. Real-time polymerase chain reaction was used to analyze the hypoxia signaling molecules' gene expression in MG-63 cells and zebrafish embryos. The findings from the scratch wound healing demonstrated that noscapine stopped MG-63 cancer cells from migrating under both hypoxia and normoxia. Blood vessel development and the heart rate in zebrafish embryos were significantly reduced by noscapine under both hypoxia and normoxia which showed the hemodynamics impact of noscapine. Noscapine also downregulated the cobalt chloride (CoCl2) induced hypoxic signaling molecules' gene expression in MG-63 cells and zebrafish embryos. Therefore, noscapine may prevent MG-63 cancer cells from proliferating and migrating, as well as decrease the formation of new vessels and the production of growth factors linked to angiogenesis in vivo under both normoxic and hypoxic conditions.


Subject(s)
Hemodynamics , Neovascularization, Pathologic , Noscapine , Zebrafish , Animals , Humans , Noscapine/pharmacology , Cell Line, Tumor , Hemodynamics/drug effects , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Hypoxia , Cell Movement/drug effects , Embryo, Nonmammalian/drug effects , Osteosarcoma/drug therapy , Angiogenesis
6.
Molecules ; 29(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38731540

ABSTRACT

Deferoxamine, an iron chelator used to treat diseases caused by excess iron, has had a Food and Drug Administration-approved status for many years. A large number of studies have confirmed that deferoxamine can reduce inflammatory response and promote angiogenesis. Blood vessels play a crucial role in sustaining vital life by facilitating the delivery of immune cells, oxygen, and nutrients, as well as eliminating waste products generated during cellular metabolism. Dysfunction in blood vessels may contribute significantly to the development of life-threatening diseases. Anti-angiogenesis therapy and pro-angiogenesis/angiogenesis strategies have been frequently recommended for various diseases. Herein, we describe the mechanism by which deferoxamine promotes angiogenesis and summarize its application in chronic wounds, bone repair, and diseases of the respiratory system. Furthermore, we discuss the drug delivery system of deferoxamine for treating various diseases, providing constructive ideas and inspiration for the development of new treatment strategies.


Subject(s)
Deferoxamine , Neovascularization, Physiologic , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Humans , Animals , Neovascularization, Physiologic/drug effects , Regeneration/drug effects , Wound Healing/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Angiogenesis
7.
J Cancer Res Clin Oncol ; 150(5): 257, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753184

ABSTRACT

PURPOSE: Breast cancer metastasis relies on cellular invasion and angiogenesis facilitated by the downregulation of metastatic suppressor proteins like Cluster of Differentiation 82 (CD82). Currently, no medicines target multiple systems to prevent metastatic progression through CD82 upregulation. This study screened for plant extracts displaying effects on cell proliferation, invasion, and CD82 expression in breast cancer cells, and in vivo angiogenesis, and further correlated between the biological activities and effect on CD82 expression. METHODS: Seventeen ethanolic plant extracts were screened for their effect on cell proliferation (against MDA-MB-231 and MCF-7 breast cancer and Hek293 kidney cells), cell invasion and effect on CD82 expression in metastatic MDA-MB-231 cells. Selected extracts were further evaluated for in vivo anti-angiogenesis. RESULTS: Extracts displayed varying antiproliferative activity against the different cell lines, and those that showed selectivity indexes (SI) > 0.5 against MDA-MB-231 were selected for anti-invasion evaluation. Buddleja saligna Willd. (BS), Combretum apiculatum Sond. (CA), Foeniculum vulgare, Greyia radlkoferi, Gunnera perpensa and Persicaria senegalensis (Meisn.) Soják (PS) displayed 50% inhibitory concentration (IC50) values of 44.46 ± 3.46, 74.00 ± 4.48, 180.43 ± 4.51, 96.97 ± 2.29, 55.29 ± 9.88 and 243.60 ± 2.69 µg/mL, respectively against MDA-MB-231, and compared to Hek293 showed SI of 0.9, 0.7, 1.4, 1.1, 2.2 and 0.5. Significant invasion inhibition was observed at both 20 and 40 µg/mL for BS (94.10 ± 0.74 and 96.73 ± 0.95%) and CA (87.42 ± 6.54 and 98.24 ± 0.63%), whereas GR (14.91 ± 1.62 and 41 ± 1.78%) and PS (36.58 ± 0.54 and 51.51 ± 0.83%), only showed significant inhibition at 40 µg/mL, and FV (< 5% inhibition) and GP (10 ± 1.03 and 22 ± 1.31%) did not show significant inhibition at both concentrations. Due to the significant anti-invasive activity of BS, CA and PS at 40 µg/mL, these extracts were further evaluated for their potential to stimulate CD82. BS showed significant (p < 0.05) reduction in CD82 at 20 and 40 µg/mL (13.2 ± 2.2% and 20.3 ± 1.5% decrease, respectively), whereas both CA and PS at 20 µg/mL increased (p < 0.05) CD82 expression (16.4 ± 0.8% and 5.4 ± 0.6% increase, respectively), and at 40 µg/mL significantly reduced CD82 expression (23.4 ± 3.1% and 11.2 ± 2.9% decrease, respectively). Using the yolk sac membrane assay, BS (59.52 ± 4.12 and 56.72 ± 3.13% newly formed vessels) and CA (83.33 ± 3.17 and 74.00 ± 2.12%) at both 20 and 40 µg/egg showed significant (p < 0.001) angiogenesis inhibition, with BS showing statistical similar activity to the positive control, combretastatin A4 (10 nmol/egg), whereas PS only displayed significant (p < 0.001) angiogenesis stimulation at 40 µg/egg (120.81 ± 3.34% newly formed vessels). CONCLUSION: BS exhibits antiproliferative, anti-invasive, and anti-angiogenic activity despite inhibiting CD82, suggesting an alternative mode of action. CA at 20 µg/mL shows moderate anti-invasive and anti-angiogenic potential by stimulating CD82, while at 40 µg/mL it still displays these properties but inhibits CD82, suggesting an additional mode of action. PS, with the least antiproliferative activity, stimulates CD82 and inhibits angiogenesis at 20 µg/mL but inhibits CD82 and increases angiogenesis at 40 µg/mL, indicating CD82 targeting as a major mode of action. Future studies should explore breast cancer xenograft models to assess the extracts' impact on CD82 expression and angiogenesis in the tumor microenvironment, along with isolating bioactive compounds from the extracts.


Subject(s)
Breast Neoplasms , Cell Proliferation , Kangai-1 Protein , Neoplasm Invasiveness , Neovascularization, Pathologic , Plant Extracts , Humans , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Plant Extracts/pharmacology , Female , Animals , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Kangai-1 Protein/metabolism , Plants, Medicinal/chemistry , HEK293 Cells , Cell Line, Tumor , Ethanol/chemistry , Ethanol/pharmacology , Chick Embryo , Neoplasm Metastasis , Chorioallantoic Membrane/drug effects , Angiogenesis
8.
J Tradit Chin Med ; 44(3): 458-467, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38767629

ABSTRACT

OBJECTIVE:To elucidate the mechanism by which Huoxue Jiedu Huayu recipe (, HJHR) regulates angiogenesis in the contralateral kidney of unilateral ureteral obstruction (UUO) rats and the mechanism by which it reduces of renal fibrosis. METHODS: Male Wistar rats were randomly divided into 4 groups: the sham group, UUO group (180 d of left ureter ligation), UUO plus eplerenone (EPL) group, and UUO plus HJHR group. After 180 d of oral drug administration, blood and contralateral kidneys were collected for analysis. Angiogenesis- and fibrosis-related indexes were detected. RESULTS: HJHR and EPL improved structural damage and renal interstitial fibrosis in the contralateral kidney and reduced the protein expression levels of α-smooth muscle actin (α-SMA), vimentin and collagen I. Moreover, these treatments could reduce the expression of vascular endothelial growth factor-A (VEGFA) by inhibiting the infiltration of macrophages. Furthermore, HJHR and EPL significantly reduced the expression of CD34 and CD105 by downregulating VEGFA production, which inhibited angiogenesis. Finally, the coexpressions of CD34, CD105 and α-SMA were decreased in the HJHR and EPL groups, indicating that endothelial-to-mesenchymal transition was inhibited. CONCLUSIONS: These findings confirm that HJHR alleviates contralateral renal fibrosis by inhibiting VEGFA-induced angiogenesis, encourage the use of HJHR against renal interstitial fibrosis and provide a theoretical basis for the clinical management of patients with CKD.


Subject(s)
Drugs, Chinese Herbal , Fibrosis , Kidney , Macrophages , Rats, Wistar , Ureteral Obstruction , Vascular Endothelial Growth Factor A , Animals , Male , Ureteral Obstruction/metabolism , Ureteral Obstruction/drug therapy , Ureteral Obstruction/genetics , Rats , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Kidney/drug effects , Kidney/metabolism , Macrophages/drug effects , Macrophages/metabolism , Drugs, Chinese Herbal/administration & dosage , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Kidney Diseases/etiology , Kidney Diseases/genetics , Angiogenesis
9.
BMC Cancer ; 24(1): 614, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773427

ABSTRACT

OBJECTIVE: Our study was to investigate the impact of taurolactone, a novel anti-tumor and anti-angiogenic drug, on AGGF1, an angiogenic factor, and angiogenesis mimicry in patients diagnosed with hepatocellular carcinoma (HCC). METHODS: A total of 120 HCC patients were enrolled from the Department of Oncology and Hepatobiliary Surgery at our hospital between May 2021 and December 2022. HCC diagnoses were confirmed through imaging or tissue biopsy for all patients. The age of patients ranged from 37 to 72 years, with an average age of 64.29 ± 4.58 years. These participants were divided equally into two groups: the control group and the observation group, each consisting of 60 individuals. While the control group received standard drug treatment, the observation group was administered taurolactone treatment. Before being included in the study, all participants or their legal representatives provided signed informed consent. Patient demographic information was collected through a questionnaire survey. ELISA was used to measure the levels of VEGF and AGGF1 in patients following treatment. Western blot was applied to assess the protein expression of PDGF, Angiopoietin, and AGGF1. MRI imaging technology was utilized to assess the perfusion characteristics of tumor blood vessels in patients. Tumor vessel density was compared between patients using ultrasonography. We also conducted a comparison between the two groups in terms of progression-free survival and overall survival. RESULTS: General patient information between the two groups showed no significant differences (P > 0.05). Of note, the observation group exhibited greatly lower levels of VEGF and AGGF1 compared to the control group (P < 0.05). Moreover, the levels of PDGF, Angiopoietin, and AGGF1 protein expression were significantly reduced in the observation group compared to the control group (P < 0.05). In terms of tumor perfusion, the observation group displayed lower average and maximum perfusion volumes in tumor blood vessels compared to the control group (P < 0.05). Additionally, the observation group demonstrated delayed peak times and arrival times of tumor blood vessels in comparison to the control group (P < 0.05). Furthermore, the density of tumor blood vessels was notably lower in the observation group compared to the control group (P < 0.05). Patients in the observation group had longer progression-free survival and overall survival than the control group (P < 0.05). CONCLUSION: In HCC patients, our study highlighted the potential efficacy of taurolactone treatment as it effectively inhibited angiogenic factors and angiogenesis mimicry, ultimately leading to an improved prognosis for these patients.


Subject(s)
Angiogenesis Inhibitors , Angiogenic Proteins , Carcinoma, Hepatocellular , Liver Neoplasms , Neovascularization, Pathologic , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Middle Aged , Male , Female , Aged , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenic Proteins/metabolism , Adult , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Lactones/therapeutic use , Lactones/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis
10.
Zhongguo Zhong Yao Za Zhi ; 49(10): 2619-2628, 2024 May.
Article in Chinese | MEDLINE | ID: mdl-38812162

ABSTRACT

Nontraumatic avascular necrosis of the femoral head(NANFH) is a common and refractory femoral head disease that causes bone death due to interruption of blood supply. Early clinical symptoms are atypical, such as hip pain and limited joint function. In the late stage, severe pain, shortening of the affected limb, claudication, and other serious symptoms are common, which se-riously affects the quality of life of patients. Therefore, it is of great significance to actively improve the clinical symptoms of NANFH to enhance the quality of life of patients. The pathogenesis of NANFH is complex, such as traumatic vascular circulatory disorders, the use of hormones or other drugs, alcoholism, and diabetes mellitus. These factors directly or indirectly lead to femoral head vascular damage, thrombosis, and coagulation system disorders, which reduce the blood supply to the acetabulum and femoral head, thus causing ischaemic death of the femoral head or even femoral head collapse. NANFH is mainly categorized as "bone impotence" and "bone paralysis" in traditional Chinese medicine(TCM). The treatment of NANFH with TCM has the characteristics and advantages of a long history, stable and reliable therapeutic effect, fewer adverse reactions, good patient tolerance, and high acceptance. Previous studies have shown that the promotion of angiogenesis is a key initiative in the prevention and treatment of NANFH, and TCM can promote fe-moral head angiogenesis by interfering with the expression of angiogenesis-related factors, which in turn can help to restore the blood supply of the femoral head and thus improve clinical symptoms of NANFH and prevent and treat NANFH. This article described the roles of blood supply interruption and angiogenesis in NANFH and the accumulated knowledge and experience of TCM in NANFH and summarized the role of angiogenesis-related factors in NANFH and the research progress on TCM intervention, so as to provide an idea for the subsequent research and a new basis for the clinical application of TCM in the treatment of NANFH.


Subject(s)
Drugs, Chinese Herbal , Femur Head Necrosis , Humans , Femur Head Necrosis/prevention & control , Femur Head Necrosis/drug therapy , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/administration & dosage , Medicine, Chinese Traditional , Animals , Femur Head/blood supply , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Angiogenesis
11.
Am J Chin Med ; 52(3): 583-604, 2024.
Article in English | MEDLINE | ID: mdl-38716616

ABSTRACT

In recent years, due to advancements in medical conditions and the development of scientific research, the fundamental research of TCM antitumor treatments has progressed from the cellular level to the molecular and genetic levels. Previous studies have demonstrated the significant role of traditional Chinese medicine (TCM) in antitumor therapy through various mechanisms and pathways. Its mechanism of action is closely associated with cancer biology across different stages. This includes inhibiting tumor cell proliferation, blocking invasion and metastasis to surrounding tissues, inducing tumor cell apoptosis, inhibiting tumor angiogenesis, regulating immune function, maintaining genome stability, preventing mutation, and regulating cell energy metabolism. The use of TCM for eliciting antitumor effects not only has a good therapeutic effect and low side effects, it also provides a solid theoretical basis for clinical treatment and medication. This paper reviews the mechanism of the antitumor effects of TCM based on tumor characteristics. Through our review, we found that TCM not only directly inhibits tumors, but also enhances the body's immunity, thereby indirectly inducing an antitumor effect. This function aligns with the TCM theory of "strengthening the body's resistance to eliminate pathogenic factors". Furthermore, TCM will play a significant role in tumor treatment in clinical settings.


Subject(s)
Apoptosis , Drugs, Chinese Herbal , Medicine, Chinese Traditional , Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Apoptosis/drug effects , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Neovascularization, Pathologic/drug therapy , Cell Proliferation/drug effects , Phytotherapy , Genomic Instability , Energy Metabolism/drug effects
12.
Int J Mol Sci ; 25(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38791428

ABSTRACT

Glioblastoma multiforme (GBM) represents the deadliest tumor among brain cancers. It is a solid tumor characterized by uncontrolled cell proliferation generating the hypoxic niches in the cancer core. By inducing the transcription of hypoxic inducible factor (HIF), hypoxia triggers many signaling cascades responsible for cancer progression and aggressiveness, including enhanced expression of vascular endothelial growth factor (VEGF) or antioxidant enzymes, such as heme oxygenase-1 (HO-1). The present work aimed to investigate the link between HO-1 expression and the hypoxic microenvironment of GBM by culturing two human glioblastoma cell lines (U87MG and A172) in the presence of a hypoxic mimetic agent, deferoxamine (DFX). By targeting hypoxia-induced HO-1, we have tested the effect of a novel acetamide-based HO-1 inhibitor (VP18/58) on GBM progression. Results have demonstrated that hypoxic conditions induced upregulation and nuclear expression of HO-1 in a cell-dependent manner related to malignant phenotype. Moreover, our data demonstrated that the HO-1 inhibitor counteracted GBM progression by modulating the HIFα/HO-1/VEGF signaling cascade in cancer cells bearing more malignant phenotypes.


Subject(s)
Acetamides , Glioblastoma , Heme Oxygenase-1 , Signal Transduction , Vascular Endothelial Growth Factor A , Humans , Glioblastoma/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Heme Oxygenase-1/metabolism , Cell Line, Tumor , Acetamides/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Signal Transduction/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Cell Proliferation/drug effects , Disease Progression , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Cell Hypoxia/drug effects
13.
Int J Mol Sci ; 25(10)2024 May 20.
Article in English | MEDLINE | ID: mdl-38791608

ABSTRACT

Due to its propensity to metastasize, cancer remains one of the leading causes of death worldwide. Thanks in part to their intrinsic low cytotoxicity, the effects of the flavonoid family in the prevention and treatment of various human cancers, both in vitro and in vivo, have received increasing attention in recent years. It is well documented that Apigenin (4',5,7-trihydroxyflavone), among other flavonoids, is able to modulate key signaling molecules involved in the initiation of cancer cell proliferation, invasion, and metastasis, including JAK/STAT, PI3K/Akt/mTOR, MAPK/ERK, NF-κB, and Wnt/ß-catenin pathways, as well as the oncogenic non-coding RNA network. Based on these premises, the aim of this review is to emphasize some of the key events through which Apigenin suppresses cancer proliferation, focusing specifically on its ability to target key molecular pathways involved in angiogenesis, epithelial-to-mesenchymal transition (EMT), maintenance of cancer stem cells (CSCs), cell cycle arrest, and cancer cell death.


Subject(s)
Apigenin , Epithelial-Mesenchymal Transition , Neoplasms , Apigenin/pharmacology , Apigenin/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Animals , Epithelial-Mesenchymal Transition/drug effects , Signal Transduction/drug effects , Cell Proliferation/drug effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism
14.
Molecules ; 29(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38792080

ABSTRACT

Tumor cells in hypoxic conditions control cancer metabolism and angiogenesis by expressing HIF-1α. Tanshinone is a traditional Chinese medicine that has been shown to possess antitumor properties and exerts a therapeutic impact on angiogenesis. However, the precise molecular mechanism responsible for the antitumor activity of 3-Hydroxytanshinone (3-HT), a type of tanshinone, has not been fully understood. Therefore, our study aimed to investigate the mechanism by which 3-HT regulates the expression of HIF-1α. Our findings demonstrate that 3-HT inhibits HIF-1α activity and expression under hypoxic conditions. Additionally, 3-HT inhibits hypoxia-induced angiogenesis by suppressing the expression of VEGF. Moreover, 3-HT was found to directly bind to α-enolase, an enzyme associated with glycolysis, resulting in the suppression of its activity. This inhibition of α-enolase activity by 3-HT leads to the blockade of the glycolytic pathway and a decrease in glycolysis products, ultimately altering HIF1-α expression. Furthermore, 3-HT negatively regulates the expression of HIF-1α by altering the phosphorylation of AMP-activated protein kinase (AMPK). Our study's findings elucidate the mechanism by which 3-HT regulates HIF-1α through the inhibition of the glycolytic enzyme α-enolase and the phosphorylation of AMPK. These results suggest that 3-HT holds promise as a potential therapeutic agent for hypoxia-related angiogenesis and tumorigenesis.


Subject(s)
Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Phosphopyruvate Hydratase , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Phosphopyruvate Hydratase/metabolism , Phosphopyruvate Hydratase/genetics , Glycolysis/drug effects , Humans , Abietanes/pharmacology , Cell Hypoxia/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism
15.
Anticancer Res ; 44(6): 2377-2392, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821582

ABSTRACT

BACKGROUND/AIM: Oral squamous cell carcinoma (OSCC) is characterized by early metastasis, clinical resistance and poor prognosis. Recently, we showed that aggressive OSCC cells co-express endothelial cell markers and can form tube-like structures, known as vasculogenic mimicry (VM), a process associated with poor prognosis in head and neck cancers. Given the limited success of current antiangiogenic therapy in treating OSCC, this study sought to explore the efficiency of these drugs in targeting an ex vivo model of VM. MATERIALS AND METHODS: OSCC cell lines from the tongue and floor of the mouth in addition to human endothelial cells were used. The treatments comprised a set of clinically relevant antiangiogenic drugs: sorafenib, sunitinib, and axitinib, which were administered in different doses. Multiple ex vivo approaches including cell tubulogenesis, proliferation, apoptosis, and migration assays were used. RESULTS: Although these drugs inhibited the formation of endothelial cell capillaries, they showed clear differential effects on OSCC cell-derived VM and cell morphology. Sorafenib inhibited the tubulogenesis of aggressive OSCC cells compared with the limited effect of sunitinib and axitinib. Furthermore, our data consistently demonstrated a preferential efficacy of certain drugs over others. Sorafenib and sunitinib exhibited anti-cancer effects on tumor cell proliferation, apoptosis, and cell migration, compared with the limited effect of axitinib. CONCLUSION: The antiangiogenic drugs, except sorafenib, had limited effect on VM formation in vitro and exhibited varying anti-cancer effects on OSCC cells. These data support the notion that VM formation may in part explain the development of drug resistance in OSCC cells.


Subject(s)
Angiogenesis Inhibitors , Axitinib , Cell Movement , Cell Proliferation , Mouth Neoplasms , Neovascularization, Pathologic , Sorafenib , Sunitinib , Humans , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Mouth Neoplasms/blood supply , Mouth Neoplasms/metabolism , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Sorafenib/pharmacology , Sorafenib/therapeutic use , Sunitinib/pharmacology , Sunitinib/therapeutic use , Cell Proliferation/drug effects , Cell Movement/drug effects , Axitinib/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/metabolism , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Niacinamide/therapeutic use
16.
Biomaterials ; 309: 122586, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38718615

ABSTRACT

It is imperative to optimize chemotherapy for heightened anti-tumor therapeutic efficacy. Unrestrained tumor cell proliferation and sustained angiogenesis are pivotal for cancer progression. Plinabulin, a vascular disrupting agent, selectively destroys tumor blood vessels. Tirapazamine (TPZ), a hypoxia-activated prodrug, intensifies cytotoxicity in diminishing oxygen levels within tumor cells. Despite completing Phase III clinical trials, both agents exhibited modest treatment efficiency due to dose-limiting toxicity. In this study, we employed methoxy poly(ethylene glycol)-b-poly(D,L-lactide) (mPEG-b-PDLLA) to co-deliver Plinabulin and TPZ to the tumor site, concurrently disrupting blood vessels and eliminating tumor cells, addressing both symptoms and the root cause of tumor progression. Plinabulin was converted into a prodrug with esterase response (PSM), and TPZ was synthesized into a hexyl chain-containing derivative (TPZHex) for effective co-delivery. PSM and TPZHex were co-encapsulated with mPEG-b-PDLLA, forming nanodrugs (PT-NPs). At the tumor site, PT-NPs responded to esterase overexpression, releasing Plinabulin, disrupting blood vessels, and causing nutritional and oxygen deficiency. TPZHex was activated in response to increased hypoxia, killing tumor cells. In treating 4T1 tumors, PT-NPs demonstrated enhanced therapeutic efficacy, achieving a 92.9 % tumor suppression rate and a 20 % cure rate. This research presented an innovative strategy to enhance synergistic efficacy and reduce toxicity in combination chemotherapy.


Subject(s)
Polyethylene Glycols , Tirapazamine , Tirapazamine/pharmacology , Animals , Cell Line, Tumor , Humans , Polyethylene Glycols/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/drug therapy , Triazines/pharmacology , Triazines/chemistry , Triazines/therapeutic use , Diketopiperazines
17.
Int Immunopharmacol ; 134: 112187, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38733825

ABSTRACT

OBJECTIVE: Glioblastoma (GBM) has poor clinical prognosis due to limited treatment options. In addition, the current treatment regimens for GBM may only slightly prolong patient survival. The aim of this study was to assess the role of BMAL1 in the immune microenvironment and drug resistance of GBM. METHODS: GBM cell lines with stable BMAL1 knockdown or LDHA overexpression were constructed, and functionally characterized by the CCK8, EdU incorporation, and transwell assays. In vivo GBM model was established in C57BL/6J mice. Flow cytometry, ELISA, immunofluorescence, and RT-qPCR were performed to detect macrophage polarization. Lactate production, pathological changes, and the expression of glycolytic proteins were analyzed by HE staining, immunohistochemistry, biochemical assays, and Western blotting. RESULTS: BMAL1 silencing inhibited the malignant characteristics, lactate production, and expression of glycolytic proteins in GBM cells, and these changes were abrogated by overexpression of LDHA or exogenous lactate supplementation. Furthermore, BMAL1 knockdown induced M1 polarization of macrophages, and inhibited M2 polarization and angiogenesis in GBM cells in conditioned media. Overexpression of LDHA or presence of exogenous lactate inhibited BMAL1-induced M1 polarization and angiogenesis. Finally, BMAL1 silencing and bevacizumab synergistically inhibited glycolysis, angiogenesis and M2 polarization, and promoted M1 polarization in vivo, thereby suppressing GBM growth. CONCLUSION: BMAL1 silencing can sensitize GBM cells to bevacizumab by promoting M1/M2 polarization through the LDHA/lactate axis.


Subject(s)
ARNTL Transcription Factors , Bevacizumab , Glioblastoma , Lactic Acid , Mice, Inbred C57BL , Animals , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Glioblastoma/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Cell Line, Tumor , Bevacizumab/therapeutic use , Bevacizumab/pharmacology , Mice , Lactic Acid/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Tumor Microenvironment/drug effects , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Glycolysis/drug effects , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/drug therapy , Gene Silencing , L-Lactate Dehydrogenase
18.
Int Immunopharmacol ; 134: 112180, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38733822

ABSTRACT

TGFBI, an extracellular matrix protein induced by transforming growth factor ß, has been found to exhibit aberrant expression in various types of cancer. TGFBI plays a crucial role in tumor cell proliferation, angiogenesis, and apoptosis. It also facilitates invasion and metastasis in various types of cancer, including colon, head and neck squamous, renal, and prostate cancers. TGFBI, a prominent p-EMT marker, strongly correlates with lymph node metastasis. TGFBI demonstrates immunosuppressive effects within the tumor immune microenvironment. Targeted therapy directed at TGFBI shows promise as a potential strategy to combat cancer. Hence, a comprehensive review was conducted to examine the impact of TGFBI on various aspects of tumor biology, including cell proliferation, angiogenesis, invasion, metastasis, apoptosis, and the immune microenvironment. This review also delved into the underlying biochemical mechanisms to enhance our understanding of the research advancements related to TGFBI in the context of tumors.


Subject(s)
Neoplasms , Transforming Growth Factor beta , Tumor Microenvironment , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Animals , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Transforming Growth Factor beta/metabolism , Molecular Targeted Therapy , Extracellular Matrix Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Apoptosis/drug effects , Cell Proliferation/drug effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
19.
Biomed Pharmacother ; 175: 116714, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761419

ABSTRACT

Cancer is one of the top 10 fatal diseases worldwide, among which advanced metastatic carcinoma has the highest mortality rate. Sunitinib and immune checkpoint blockers are commonly used to treat metastatic renal carcinoma with limited efficacy. Therefore, there is an urgent need to develop novel targeted therapies for metastatic renal cancer. In this study, we designed an antibody fusion protein, 57103, that simultaneously targeted the cluster of differentiation 24 (CD24), interleukin 4 receptor (IL-4R), and integrin receptors αvß3 and α5ß1. In vitro assays showed that 57103 significantly suppressed the proliferation, migration, invasion, colony formation, and adhesion abilities of renal cancer cells, resulting in a comprehensive and significant antitumor effect. Furthermore, 57103 inhibited angiogenesis, promoted THP1-derived M0-type macrophage phagocytosis, and enhanced the antibody-dependent cellular cytotoxicity of peripheral blood mononuclear and NK92MI-CD16a cells. In vivo experiments revealed significant inhibition of tumor growth in ACHN cell xenograft nude mice and an MC38-hCD24 tumor-bearing mouse model. Immunohistochemical analysis showed that 57103 decreased the proliferation and induced the apoptosis of renal cancer cells, while inhibiting angiogenesis. The MC38-hPDL1 and MC38-hCD24-hPDL1 tumor-bearing mouse models further offer the possibility of combining 57103 with the PDL1 antagonist atezolizumab. In conclusion, 57103 is a potential candidate drug for the treatment of metastatic renal carcinoma or PDL1-overexpressing cancer.


Subject(s)
Cell Proliferation , Integrin alphaVbeta3 , Kidney Neoplasms , Mice, Nude , Tumor Microenvironment , Animals , Humans , Tumor Microenvironment/drug effects , Cell Line, Tumor , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Mice , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Recombinant Fusion Proteins/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Apoptosis/drug effects , Mice, Inbred BALB C , Cell Movement/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
20.
Nanoscale ; 16(21): 10350-10365, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38739006

ABSTRACT

Tumour cells secrete various proangiogenic factors like VEGF, PDGF, and EGF that result in the formation of highly vascularized tumours with an immunosuppressive tumour microenvironment. As tumour growth and metastasis are highly dependent on angiogenesis, targeting tumour vasculature along with rapidly dividing tumour cells is a potential approach for cancer treatment. Here, we specifically engineered sub-100 sized nanomicelles (DTX-CA4 NMs) targeting proliferation and angiogenesis using an esterase-sensitive phosphocholine-tethered docetaxel conjugate of lithocholic acid (LCA) (PC-LCA-DTX) and a poly(ethylene glycol) (PEG) derivative of an LCA-combretastatin A4 conjugate (PEG-LCA-CA4). DTX-CA4 NMs effectively inhibit the tumour growth in syngeneic (CT26) and xenograft (HCT116) colorectal cancer models, inhibit tumour recurrence, and enhance the percentage survival in comparison with individual drug-loaded NMs. DTX-CA4 NMs enhance the T cell-mediated anti-tumour immune response and DTX-CA4 NMs in combination with an immune checkpoint inhibitor, anti-PDL1 antibody, enhance the anti-tumour response. We additionally showed that DTX-CA4 NMs effectively attenuate the production of ceramide-1-phosphate, a key metabolite of the sphingolipid pathway, by downregulating the expression of ceramide kinase at both transcriptional and translational levels. Therefore, this study presents the engineering of effective DTX-CA4 NMs for targeting the tumour microenvironment that can be explored further for clinical applications.


Subject(s)
Cell Proliferation , Ceramides , Docetaxel , Micelles , Neovascularization, Pathologic , Animals , Ceramides/chemistry , Ceramides/pharmacology , Humans , Mice , Cell Proliferation/drug effects , Docetaxel/pharmacology , Docetaxel/chemistry , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Lithocholic Acid/chemistry , Lithocholic Acid/pharmacology , Polyethylene Glycols/chemistry , Cell Line, Tumor , Mice, Inbred BALB C , Stilbenes/chemistry , Stilbenes/pharmacology , HCT116 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Female , Angiogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...