Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 398
1.
Mol Med ; 30(1): 57, 2024 May 02.
Article En | MEDLINE | ID: mdl-38698308

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.


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
2.
Int. j. morphol ; 40(6): 1587-1593, dic. 2022. ilus, tab, graf
Article En | LILACS | ID: biblio-1421824

SUMMARY: This study is to investigate the role and mechanism of RGD peptide in laryngeal cancer stem cells (CSCs). Laryngeal cancer CD133+Hep-2 CSCs were sorted by flow cytometry. RGD peptide was co-cultured with sorted laryngeal CSCs. Cell proliferation was detected with CCK-8 assay. The mRNA levels of VEGF/VEGFR2/STAT 3/HIF-1α were detected with RT-PCR. The proteins of VEGF/ VEGFR2/STAT 3/HIF-1α were detected with Western blot. The sorted CSCs were inoculated into nude mice. Tumor volume was measured. Integrin αvβ3 expression in tumor tissues was analyzed with immunohistochemistry. The results showed that the ratio of CD133+ CSCs to the total number of cells was 1.34±0.87 %, while CD133-non-tumor stem cells accounted for 95.0±5.76 %. The sorted cancer stem cells grew well. The RGD peptide significantly inhibited the proliferation of CD133+Hep-2 laryngeal CSCs in a dose-dependent manner. The RGD peptide significantly inhibited the mRNA of VEGFR2, STAT3 and HIF-1α in laryngeal CSCs in a concentration-dependent manner. Consistently, the RGD peptide significantly inhibited the protein expression of VEGFR2, STAT3 and HIF-1α in laryngeal CSCs in a dose-dependent manner. At the same time, in vivo tumor experiments showed that the RGD peptide significantly inhibited tumor volume but not the body weight. Furthermore, RGD peptide significantly inhibited the expression of tumor angiogenesis-related protein integrin αvβ3. Our findings demonstrate that RGD peptide inhibits tumor cell proliferation and tumor growth. The underlying mechanism may that RGD inhibits tumor angiogenesis-related signaling pathways, thus affecting the tumor angiogenesis, and decreasing the progression of human laryngeal CSCs.


Este estudio se realizó para investigar el papel y el mecanismo del péptido RGD en las células madre del cáncer de laringe (CSC). Las CSC CD133+Hep-2 de cáncer de laringe se clasificaron mediante citometría de flujo. El péptido RGD se cocultivó con CSC laríngeas clasificadas. La proliferación celular se detectó con el ensayo CCK-8. Los niveles de ARNm de VEGF/VEGFR2/ STAT 3/HIF-1α se detectaron con RT-PCR. Las proteínas de VEGF/ VEGFR2/STAT 3/HIF-1α se detectaron con Western blot. Las CSC clasificadas se inocularon en ratones nudos. Se midió el volumen del tumor. La expresión de integrina αvβ3 en tejidos tumorales se analizó con inmunohistoquímica. Los resultados mostraron que la proporción de CSC CD133+ con respecto al número total de células fue de 1,34 ± 0,87 %, mientras que las células madre no tumorales CD133 representaron el 95,0 ± 5,76 %. Las células madre cancerosas clasificadas crecieron bien. El péptido RGD inhibió significativamente la proliferación de CSC laríngeas CD133+Hep-2 de una manera dependiente de la dosis. El péptido RGD inhibió significativamente el ARNm de VEGFR2, STAT3 y HIF-1α en CSC laríngeas de manera dependiente de la concentración. De manera consistente, el péptido RGD inhibió significativamente la expresión proteica de VEGFR2, STAT3 y HIF-1α en CSC laríngeas, de manera dependiente de la dosis. Al mismo tiempo, los experimentos con tumores in vivo mostraron que el péptido RGD inhibía significativamente el volumen del tumor pero no el peso corporal. Además, el péptido RGD inhibió significativamente la expresión de la proteína integrina αvβ3 relacionada con la angiogénesis tumoral. Nuestros hallazgos demuestran que el péptido RGD inhibe la proliferación de células tumorales y el crecimiento tumoral. El mecanismo subyacente puede ser que RGD inhiba las vías de señalización relacionadas con la angiogénesis tumoral, afectando así la angiogénesis tumoral y disminuyendo la progresión de las CSC laríngeas humanas.


Animals , Mice , Oligopeptides/metabolism , Neoplastic Stem Cells , Laryngeal Neoplasms , RNA, Messenger/antagonists & inhibitors , Immunohistochemistry , Blotting, Western , DNA Primers , Reverse Transcriptase Polymerase Chain Reaction , Integrin alphaVbeta3/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Cell Proliferation , Flow Cytometry , Neovascularization, Pathologic
3.
Bioengineered ; 13(2): 4557-4572, 2022 02.
Article En | MEDLINE | ID: mdl-35142593

Integrins play an important role in multiple stages of tumor progression and metastasis. Previous studies have shown synergistic effects of combined αvß6-integrin and αvß8-integrin inhibitors with immunotherapy. However, the role of αvß3-integrin inhibitor in tumor immunity is still unclear. In this study, we aimed to elucidate the impact of the αvß3-integrin inhibitor on PD-L1 expression and sensitivity to immune checkpoint blockade in melanoma. We investigated the effects of cilengitide, an αvß3-integrin inhibitor, on cell viability and apoptosis of melanoma cell lines. And we explored how cilengitide regulated the expression of PD-L1 in melanoma cells in vitro and in vivo, using immunofluorescence, flow cytometry, Western blotting, and immunohistochemistry. A subcutaneous B16 murine melanoma model was utilized to determine whether combining cilengitide with anti-PD1 therapy inhibited tumor growth and positively regulated tumor microenvironment (TME). Our results showed that cilengitide inhibited cell viability and induced apoptosis in B16 and A375 cell lines. Furthermore, cilengitide decreased PD-L1 expression by reducing STAT3 phosphorylation in two melanoma cell lines. Cilengitide also reduced subcutaneous tumor PD-L1 expression in the B16 murine melanoma model. Accordingly, cilengitide positively regulated antitumor immune responses and provided durable therapy when combined with anti-PD1 monoclonal antibody in the murine melanoma model. This combination therapy reduced tumor growth and extended survival. Our study highlights that cilengitide enhances the efficacy of anti-PD1 therapy and produces a stronger antitumor immune response. This combination therefore represents a novel therapeutic regimen that may improve immunotherapy treratment.


B7-H1 Antigen/antagonists & inhibitors , Integrin alphaVbeta3/antagonists & inhibitors , Melanoma, Experimental/metabolism , Skin Neoplasms/metabolism , Snake Venoms/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL
4.
Int J Mol Sci ; 23(3)2022 Feb 03.
Article En | MEDLINE | ID: mdl-35163668

Breast cancer is characterized by a hypoxic microenvironment inside the tumor mass, contributing to cell metastatic behavior. Hypoxia induces the expression of hypoxia-inducible factor (HIF-1α), a transcription factor for genes involved in angiogenesis and metastatic behavior, including the vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs), and integrins. Integrin receptors play a key role in cell adhesion and migration, being considered targets for metastasis prevention. We investigated the migratory behavior of hypoxia-cultured triple-negative breast cancer cells (TNBC) and endothelial cells (HUVEC) upon αvß3 integrin blocking with DisBa-01, an RGD disintegrin with high affinity to this integrin. Boyden chamber, HUVEC transmigration, and wound healing assays in the presence of DisBa-01 were performed in hypoxic conditions. DisBa-01 produced similar effects in the two oxygen conditions in the Boyden chamber and transmigration assays. In the wound healing assay, hypoxia abolished DisBa-01's inhibitory effect on cell motility and decreased the MMP-9 activity of conditioned media. These results indicate that αvß3 integrin function in cell motility depends on the assay and oxygen levels, and higher inhibitor concentrations may be necessary to achieve the same inhibitory effect as in normoxia. These versatile responses add more complexity to the role of the αvß3 integrin during tumor progression.


Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endothelial Cells/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Tumor Hypoxia , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Shape/drug effects , Crotalid Venoms/pharmacology , Culture Media, Conditioned/pharmacology , Disintegrins/pharmacology , Endothelial Cells/pathology , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Matrix Metalloproteinases/metabolism , Neovascularization, Physiologic/drug effects , Oxygen , Protein Subunits/metabolism , Tumor Hypoxia/drug effects
5.
J Nanobiotechnology ; 19(1): 446, 2021 Dec 23.
Article En | MEDLINE | ID: mdl-34949198

The integrin αvß3 receptor and Lactoferrin receptor (LfR) are over-expressed in both cerebral microvascular endothelial cells and glioma cells. RGD tripeptide and Lf can specifically bind with integrin αvß3 receptor and LfR, respectively. In our study, RGD and Lf dual-modified liposomes loaded with docetaxel (DTX) were designed to enhance the brain targeting effect and treatment of glioma. Our in vitro studies have shown that RGD-Lf-LP can significantly enhance the cellular uptake of U87 MG cells and human cerebral microvascular endothelial cells (hCMEC/D3) when compared to RGD modified liposomes (RGD-LP) and Lf modified liposomes (Lf-LP). Free RGD and Lf competitively reduced the cellular uptake of RGD-Lf-LP, in particular, free RGD played a main inhibitory effect on cellular uptake of RGD-Lf-LP in U87 MG cells, yet free Lf played a main inhibitory effect on cellular uptake of RGD-Lf-LP in hCMEC/D3 cells. RGD-Lf-LP can also significantly increase penetration of U87 MG tumor spheroids, and RGD modification plays a dominating role on promoting the penetration of U87 MG tumor spheroids. The results of in vitro BBB model were shown that RGD-Lf-LP-C6 obviously increased the transport of hCMEC/D3 cell monolayers, and Lf modification plays a dominating role on increasing the transport of hCMEC/D3 cell monolayers. In vivo imaging proved that RGD-Lf-LP shows stronger targeting effects for brain orthotopic gliomas than that of RGD-LP and Lf-LP. The result of tissue distribution confirmed that RGD-LF-LP-DTX could significantly increase brain targeting after intravenous injection. Furthermore, RGD-LF-LP-DTX (a dose of 5 mg kg-1 DTX) could significantly prolong the survival time of orthotopic glioma-bearing mice. In summary, RGD and LF dual modification are good combination for brain targeting delivery, RGD-Lf-LP-DTX could enhance brain targeting effects, and is thus a promising chemotherapeutic drug delivery system for treatment of glioma.


Antineoplastic Agents/pharmacology , Docetaxel/chemistry , Integrin alphaVbeta3/antagonists & inhibitors , Liposomes/chemistry , Receptors, Cell Surface/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel/metabolism , Docetaxel/pharmacology , Docetaxel/therapeutic use , Glioma/diagnostic imaging , Glioma/drug therapy , Glioma/pathology , Humans , Integrin alphaVbeta3/metabolism , Liposomes/pharmacokinetics , Mice , Mice, Nude , Oligopeptides/chemistry , Particle Size , Receptors, Cell Surface/metabolism , Survival Rate , Tissue Distribution
6.
Scand Cardiovasc J ; 55(5): 287-296, 2021 Oct.
Article En | MEDLINE | ID: mdl-34296634

OBJECTIVE: The present study aimed to evaluate the protective role of cilengitide (CGT), an integrin αvß3 and αvß5 inhibitor, on doxorubicin (DOX)-induced myocardial fibrosis and cardiac dysfunction in a rat model. Methods. Forty male rats were randomly divided into four groups: DOX (n = 12), intraperitoneal (i.p.) injection of DOX 0.8 ∼ 1.0 mg/kg three times a week for up to 6 weeks, then saline i.p. three times a week for another 3 weeks; CGT (n = 8), CGT 10 mg/kg, i.p. three times a week for 9 weeks; DOX + CGT (n = 12), DOX and CGT co-administration as above for 6 weeks, then CGT alone for another 3 weeks; Control (n = 8), saline i.p. three times a week for 9 weeks. Echocardiography, serum procollagen I C-terminal propeptide (PICP) procollagen III N-terminal propeptide (PIIINP) and C telopeptide type I (CTX-I) were evaluated at baseline and 3, 6 and 9 weeks after initial DOX administration for all surviving rats. The heart tissues were then harvested for myocardial hydroxyproline (HYP) evaluation, qRT-PCR, and western blotting. Results. CGT attenuated DOX-induced eccentric remodeling by improving relative wall thickness at the 9th week. CGT also improved systolic function at the 9th week and diastolic function at the 6th and 9th week. CGT reduced myocardial HYP and serum PICP, PIIINP, CTX-I, and the PICP/PIIINP ratio. RT-PCR and western blot showed that CGT blocked the TGF-ß1/SMAD3 pathway and mitigating extracellular matrix turnover. Conclusions. CGT exerted a cardioprotective effect against doxorubicin-induced fibrosis and improved cardiac function.


Cardiomyopathies , Integrin alphaVbeta3 , Receptors, Vitronectin , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/epidemiology , Disease Models, Animal , Doxorubicin/toxicity , Fibrosis/prevention & control , Integrin alphaVbeta3/antagonists & inhibitors , Male , Rats , Receptors, Vitronectin/antagonists & inhibitors
7.
Cancer Treat Res Commun ; 28: 100395, 2021.
Article En | MEDLINE | ID: mdl-34034044

Integrins are a family of heterodimeric plasma membrane glycoproteins, which regulate tumor growth, angiogenesis, migration, and metastasis. Integrin αvß3 has been recognized as a putative target for the treatment of several cancers. Thus, the characterization of αvß3 distribution in different human tumors is of substantial interest in tumor targeting and its suppression. In this study we evaluated the expression of integrin αvß3 in different cancer types to define the expression pattern in cancer model. Furthermore, we investigated the effect of novel αvß3 antagonist Diaminopropane Tetraiodothyroacetic acid conjugated to poly (lactic-co-glycolic acid) polymer and its nanoformulated form (NDAT), on different cancer cell lines both in vitro and in xenografts. In vitro, NDAT downregulated αv and ß3 monomer expression. In vivo in tumor xenografts, similarly, NDAT downregulated αv and ß3. Distinct reduction in tumor weight and viability was observed in glioblastoma xenografts treated with NDAT. Furthermore, NDAT was safe and tolerable in mice treated with high doses. In conclusion, NDAT is an effective and safe inhibitor of integrin αvß3 expression in various cancer types, which indicates its impact on the targetability and suppression of αvß3-associated tumor functions.


Antineoplastic Agents/administration & dosage , Integrin alphaVbeta3/antagonists & inhibitors , Nanoparticles/administration & dosage , Neoplasms/drug therapy , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Thyroxine/analogs & derivatives , Animals , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin alphaVbeta3/genetics , Male , Mice, Nude , Neoplasms/genetics , Thyroxine/administration & dosage , Treatment Outcome
8.
J Med Chem ; 64(9): 6300-6309, 2021 05 13.
Article En | MEDLINE | ID: mdl-33886292

We have previously reported that the αvß3 inhibitor P-bi-TAT, a bifunctional version of the thyroid hormone metabolite tetraiodothyroacetic acid (tetrac) conjugated to polyethylene glycol (PEG) MW 4000, has excellent efficacy in a glioblastoma multiforme (GBM) mouse model. However, bioanalysis problems due to PEG polydispersity and large-scale synthesis issues led to a search for new molecules, culminating in the discovery of fb-PMT, a conjugate of tetrac and monodisperse PEG36, with a lipophilic 4-fluorobenzyl group at the opposite end of the PEG chain. fb-PMT reduces GBM tumor growth and viability by up to 98%, is suitable for large-scale synthesis, and is amenable to bioanalysis using mass spectrometry-based detection. We also showed that changes in lipophilicity at the opposite end of the PEG chain from the active tetrac component affected the proton NMR chemical shift of the tetrac moiety in D20 and brain levels of the compound after subcutaneous dosing.


Acetic Acid/chemistry , Acetic Acid/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Brain/metabolism , Glioblastoma/pathology , Integrin alphaVbeta3/antagonists & inhibitors , Acetic Acid/chemical synthesis , Acetic Acid/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemistry Techniques, Synthetic , Glioblastoma/drug therapy , Humans , Mice , Polyethylene Glycols/chemistry
9.
Chem Commun (Camb) ; 57(27): 3407-3410, 2021 Apr 07.
Article En | MEDLINE | ID: mdl-33687395

A new convergent chemoenzymatic synthesis strategy, integrating enzymatic synthesis of heparan sulfate, sortase A ligation, copper(i)-catalyzed alkyne-azide cycloaddition, and solid phase peptide synthesis, has been established to efficiently synthesize a mimetic of heparan sulfate proteoglycan syndecan-1 glyco-polypeptide at a milligram scale. The mimic was able to bind with αvß3 integrin faster and exhibit stronger inhibition of breast cancer cell migration compared to the glycan or the polypeptide alone. This novel approach could serve as a general approach for heparan sulfate proteoglycan mimetic synthesis.


Heparitin Sulfate/pharmacology , Proteoglycans/pharmacology , Syndecan-1/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Female , Heparitin Sulfate/chemistry , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Molecular Conformation , Proteoglycans/chemistry , Syndecan-1/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
10.
Cell Prolif ; 54(4): e13012, 2021 Apr.
Article En | MEDLINE | ID: mdl-33656760

OBJECTIVES: Vitronectin (VTN) has been widely used for the maintenance and expansion of human pluripotent stem cells (hPSCs) as feeder-free conditions. However, the effect of VTN on hPSC differentiation remains unclear. Here, we investigated the role of VTN in early haematopoietic development of hPSCs. MATERIALS AND METHODS: A chemically defined monolayer system was applied to study the role of different matrix or basement membrane proteins in haematopoietic development of hPSCs. The role of integrin signalling in VTN-mediated haematopoietic differentiation was investigated by integrin antagonists. Finally, small interfering RNA was used to knock down integrin gene expression in differentiated cells. RESULTS: We found that the haematopoietic differentiation of hPSCs on VTN was far more efficient than that on Matrigel that is also often used for hPSC culture. VTN promoted the fate determination of endothelial-haematopoietic lineage during mesoderm development to generate haemogenic endothelium (HE). Moreover, we demonstrated that the signals through αvß3 and αvß5 integrins were required for VTN-promoted haematopoietic differentiation. Blocking αvß3 and αvß5 integrins by the integrin antagonists impaired the development of HE, but not endothelial-to-haematopoietic transition (EHT). Finally, both αvß3 and αvß5 were confirmed acting synergistically for early haematopoietic differentiation by knockdown the expression of αv, ß3 or ß5. CONCLUSION: The established VTN-based monolayer system of haematopoietic differentiation of hPSCs presents a valuable platform for further investigating niche signals involved in human haematopoietic development.


Cell Differentiation/drug effects , Integrin alphaVbeta3/metabolism , Receptors, Vitronectin/metabolism , Vitronectin/pharmacology , Cell Adhesion/drug effects , Cell Line , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/genetics , Mesoderm/cytology , Mesoderm/growth & development , Mesoderm/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Vitronectin/antagonists & inhibitors , Receptors, Vitronectin/genetics , Signal Transduction/drug effects , Snake Venoms/pharmacology
11.
J Exp Med ; 218(4)2021 04 05.
Article En | MEDLINE | ID: mdl-33561195

Fibrotic tumor stroma plays an important role in facilitating triple-negative breast cancer (TNBC) progression and chemotherapeutic resistance. We previously reported a rationally designed protein (ProAgio) that targets integrin αvß3 at a novel site. ProAgio induces apoptosis via the integrin. Cancer-associated fibroblasts (CAFs) and angiogenic endothelial cells (aECs) in TNBC tumor express high levels of integrin αvß3. ProAgio effectively induces apoptosis in CAFs and aECs. The depletion of CAFs by ProAgio reduces intratumoral collagen and decreases growth factors released from CAFs in the tumor, resulting in decreased cancer cell proliferation and apoptotic resistance. ProAgio also eliminates leaky tumor angiogenic vessels, which consequently reduces tumor hypoxia and improves drug delivery. The depletion of CAFs and reduction in hypoxia by ProAgio decreases lysyl oxidase (LOX) secretion, which may play a role in the reduction of metastasis. ProAgio stand-alone or in combination with a chemotherapeutic agent provides survival benefit in TNBC murine models, highlighting the therapeutic potential of ProAgio as a treatment strategy.


Angiogenesis Inhibitors/pharmacology , Cancer-Associated Fibroblasts/drug effects , Neovascularization, Pathologic/drug therapy , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/drug therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Doxorubicin/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Hypoxia/drug therapy , Integrin alphaVbeta3/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/metabolism , Paclitaxel/therapeutic use , Protein-Lysine 6-Oxidase/metabolism , Signal Transduction/drug effects , Treatment Outcome , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Cell Mol Gastroenterol Hepatol ; 11(1): 161-179, 2021.
Article En | MEDLINE | ID: mdl-32810598

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is resistant to most therapeutics owing to dense fibrotic stroma orchestrated by cancer-associated pancreatic stellate cells (CAPaSC). CAPaSC also support cancer cell growth, metastasis, and resistance to apoptosis. Currently, there is no effective therapy for PDAC that specifically targets CAPaSC. We previously reported a rationally designed protein, ProAgio, that targets integrin αvß3 at a novel site and induces apoptosis in integrin αvß3-expressing cells. Because both CAPaSC and angiogenic endothelial cells express high levels of integrin αvß3, we aimed to analyze the effects of ProAgio in PDAC tumor. METHODS: Expression of integrin αvß3 was examined in both patient tissue and cultured cells. The effects of ProAgio on CAPaSC were analyzed using an apoptosis assay kit. The effects of ProAgio in PDAC tumor were studied in 3 murine tumor models: subcutaneous xenograft, genetic engineered (KrasG12D; p53R172H; Pdx1-Cre, GEM-KPC) mice, and an orthotopic KrasG12D; p53R172H; Pdx1-Cre (KPC) model. RESULTS: ProAgio induces apoptosis in CAPaSC. ProAgio treatment significantly prolonged survival of a genetically engineered mouse-KPC and orthotopic KPC mice alone or in combination with gemcitabine (Gem). ProAgio specifically induced apoptosis in CAPaSC, resorbed collagen, and opened collapsed tumor vessels without an increase in angiogenesis in PDAC tumor, enabling drug delivery into the tumor. ProAgio decreased intratumoral insulin-like growth factor 1 levels as a result of depletion of CAPaSC and consequently decreased cytidine deaminase, a Gem metabolism enzyme in cancer cells, and thereby reduced resistance to Gem-induced apoptosis. CONCLUSIONS: Our study suggests that ProAgio is an effective PDAC treatment agent because it specifically depletes CAPaSC and eliminates tumor angiogenesis, thereby enhancing drug delivery and Gem efficacy in PDAC tumors.


Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Integrin alphaVbeta3/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Models, Animal , Humans , Integrin alphaVbeta3/analysis , Integrin alphaVbeta3/metabolism , Mice , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Primary Cell Culture , Xenograft Model Antitumor Assays
13.
PLoS One ; 15(8): e0232917, 2020.
Article En | MEDLINE | ID: mdl-32810161

In human lung cancer progression, the EMT process is characterized by the transformation of cancer cells into invasive forms that migrate to other organs. Targeting to EMT-related molecules is emerging as a novel therapeutic approach for the prevention of lung cancer cell migration and invasion. Traf2- and Nck-interacting kinase (TNIK) has recently been considered as an anti-proliferative target molecule to regulate the Wnt signaling pathway in several types of cancer cells. In the present study, we evaluated the inhibitory effect of a tyrosine kinase inhibitor sunitinib and the integrin-αⅤß3 targeted cyclic peptide (cRGDfK) on EMT in human lung cancer cells. Sunitinib strongly inhibited the TGF-ß1-activated EMT through suppression of Wnt signaling, Smad and non-Smad signaling pathways. In addition, the cRGDfK also inhibited the expression of TGFß1-induced mesenchymal marker genes and proteins. The anti-EMT effect of sunitinib was enhanced when cRGDfK was treated together. When sunitinib was treated with cRGDfK, the mRNA and protein expression levels of mesenchymal markers were decreased compared to the treatment with sunitinib alone. Co-treatment of cRGDfK has shown the potential to improve the efficacy of anticancer agents in combination with therapeutic agents that may be toxic at high concentrations. These results provide new and improved therapies for treating and preventing EMT-related disorders, such as lung fibrosis and cancer metastasis, and relapse.


Carcinoma, Non-Small-Cell Lung/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/drug therapy , Peptides, Cyclic/administration & dosage , Sunitinib/administration & dosage , Transforming Growth Factor beta1/antagonists & inhibitors , A549 Cells , Adenosine Triphosphate/metabolism , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Catalytic Domain , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Docking Simulation , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Smad Proteins/metabolism , Wnt Signaling Pathway/drug effects
14.
J Med Chem ; 63(14): 7653-7662, 2020 07 23.
Article En | MEDLINE | ID: mdl-32571015

Therapeutic targeting of the norepinephrine transporter (NET) function with benzylguanidine (BG), conjugated with the high-affinity thyrointegrin αvß3 antagonist triazole tetraiodothyroacetic acid, TAT, via noncleavable bonding to poly(ethylene glycol) (PEG400) (P) might allow for effective treatment options in neuroblastoma. BG-P-TAT is a dual-targeting agent, targeting the NET function and the thyrointegrin αvß3 receptors that are overexpressed in neuroblastoma and other neuroendocrine tumors. Various cancer cells and actively dividing tumor-endothelial cells express the thyrointegrin αvß3 receptors. In this work, the novel compound BG-P-TAT was synthesized and evaluated in the neuroblastoma SK-N-FI cell line for improved targeting and to offer a new strategy for patients with neuroblastoma. BG-P-TAT demonstrated significant suppression of neuroblastoma tumor progression, growth, and viability in a dose-dependent manner. In conclusion, BG-P-TAT represents a potential lead candidate for the treatment of neuroblastoma and other neuroendocrine tumors.


Antineoplastic Agents/therapeutic use , Integrin alphaVbeta3/antagonists & inhibitors , Neuroblastoma/drug therapy , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Female , Guanidines/chemical synthesis , Guanidines/therapeutic use , Humans , Mice, Nude , Necrosis/chemically induced , Thyroxine/analogs & derivatives , Thyroxine/therapeutic use , Triazoles/chemical synthesis , Triazoles/therapeutic use , Xenograft Model Antitumor Assays
15.
Exp Dermatol ; 29(6): 562-569, 2020 06.
Article En | MEDLINE | ID: mdl-32298492

Melanoma is a deadly type of skin cancer that is particularly difficult to treat owing to its resistance to radiation therapy. Here, we attempted to determine the key proteins responsible for melanoma radioresistance, with the aim of improving disease response to radiation therapy. Two melanoma cell lines, SK-Mel5 and SK-Mel28, with different radiosensitivities were analysed via RNA-Seq (Quant-Seq) and target proteins with higher abundance in the more radioresistant cell line, SK-Mel28, identified. Among these proteins, integrin αvß3, a well-known molecule in cell adhesion, was selected for analysis. Treatment of SK-Mel28 cells with cilengitide, an integrin αvß3 inhibitor, as well as γ-irradiation resulted in more significant cell death than γ-irradiation alone. In addition, Akt, a downstream signal transducer of integrin αvß3, showed high basic activation in SK-Mel28 and was significantly decreased upon co-treatment with cilengitide and γ-irradiation. MK-2206, an Akt inhibitor, exerted similar effects on the SK-Mel28 cell line following γ-irradiation. Our results collectively demonstrate that the integrin αvß3-Akt signalling pathway contributes to radioresistance in SK-Mel28 cells, which may be manipulated to improve therapeutic options for melanoma.


Integrin alphaVbeta3/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance , Skin Neoplasms/metabolism , Cell Line, Tumor , Cell Survival , Gamma Rays , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Melanoma/radiotherapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , Skin Neoplasms/radiotherapy , Snake Venoms/pharmacology
16.
Nanoscale ; 12(13): 6953-6958, 2020 Apr 03.
Article En | MEDLINE | ID: mdl-32191787

Developing novel small-molecule-based probes with both deep tissue imaging and therapeutic functions is highly significant in cancer diagnosis and treatment. Herein, we report a novel second near-infrared (NIR-II) fluorescent probe QT-RGD constructed with a NIR-II emissive organic fluorophore and two cyclic-(arginine-glycine-aspartic acid) (cRGD) peptides that can specifically bind to the tumor-associated αvß3 integrin for accurate tumor diagnosis and targeting therapy. The isotopic 125I-labeled probe exhibited great tumor targeting ability and emitted intensive NIR-II/photoacoustic (PA)/single-photon emission computed tomography (SPECT) signals, which allows specific and sensitive multimodal visualization of tumors in vivo. More notably, this probe could also be applied for effective imaging-guided photothermal therapy (PTT) of tumors in mouse models owing to its prominent photothermal conversion efficiency and excellent photothermal stability. We thus envision that our work which unveils a combination of NIR-II/PA/SPECT imaging and PTT would offer a valuable means of improving tumor diagnostic accuracy as well as therapeutic efficacy.


Integrin alphaVbeta3/antagonists & inhibitors , Nanoparticles , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental , Optical Imaging , Photothermal Therapy , Single Photon Emission Computed Tomography Computed Tomography , Theranostic Nanomedicine , Animals , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasm Proteins/metabolism , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/therapy , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology
17.
Bioorg Med Chem Lett ; 30(8): 127039, 2020 04 15.
Article En | MEDLINE | ID: mdl-32094009

In this letter, we report a series of five new RGD-containing cyclic peptides as potent inhibitors to αvß3 integrin protein. We have incorporated various unnatural lipophilic amino acids into the cyclic RGD framework of cilengitide, which is selective for αvß3 integrin. All the newly synthesized cyclic peptides were evaluated in vitrosolid phase binding assay and investigated for their bindingbehaviourtowards integrin subtypes. All the cyclic peptides were synthesized in excellent yield following solution-phase coupling strategy. The cyclic RGD peptides 1a-e exhibited IC50 of 9.9, 5.5, 72, 11 and 3.3 nM, respectively, towardsαvß3 integrin protein. This finding offers further opportunities for the introduction unusual amino acids into the cyclic peptide framework of cilengitide.


Integrin alphaVbeta3/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Dose-Response Relationship, Drug , Humans , Integrin alphaVbeta3/metabolism , Molecular Structure , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Structure-Activity Relationship
19.
Curr Cancer Drug Targets ; 20(4): 306-315, 2020.
Article En | MEDLINE | ID: mdl-31893992

BACKGROUND: Integrins are crucial anti-cancer therapy targets. We previously showed that tablysin-15 is an integrin antagonist with its Arg-Gly-Asp motif in a novel structural context. OBJECTIVE: Here we investigated the anti-cancer effects and mechanisms of action of tablysin-15 in melanoma cells. METHODS: Cell adhesion, competitive binding, cell viability, and ATP chemiluminescence assays were used to analyze the binding of tablysin-15 to αvß3 integrin and its phenotypic effects. Wound healing, transwells, and zymography were performed to detect motility and matrix metalloproteinase- 2/-9 activities. PARP and caspase-3 cleavage were used as apoptosis assays, while LDH release and flow cytometry were used for necrosis and cell cycle analysis. The expression of mRNAs and proteins of target molecules was measured by qRT-PCR and western blotting, respectively. RESULTS: Tablysin-15 dose-dependently inhibited the proliferation, migration, and invasion of M21 cells through integrin αvß3. The proliferation inhibition caused by tablysin-15 was attributable to G0/G1 phase arrest rather than apoptosis or necrosis. Furthermore, tablysin-15 suppressed MMP-2/- 9 activities and the mRNA expression of MMP-2/-9 and COX-2 but was upregulated TIMP-1 in M21 cells. Meanwhile, tablysin-15 suppressed the expression of cyclin D1/E and CDK 2/6, the phosphorylation of FAK, Akt, and ERK, and nuclear translocation of NF-κB, while increasing the expression of the CDK inhibitor p21waf1/C1. Taken together, tablysin-15 might inhibit melanoma cell metastasis and proliferation by competing with αvß3 integrin, thereby blocking FAK-associated signaling pathways and nuclear translocation of NF-κB. CONCLUSION: Tablysin-15 has reliable anti-cancer effects against M21 melanoma cells, suggesting tablysin-15 is a promising anti-tumor drug.


Insect Proteins/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , NF-kappa B/metabolism , Salivary Proteins and Peptides/pharmacology , Skin Neoplasms/drug therapy , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Disintegrins/chemistry , Disintegrins/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Melanoma/metabolism , Melanoma/pathology , Oligopeptides/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
20.
Mol Pharm ; 17(1): 349-358, 2020 01 06.
Article En | MEDLINE | ID: mdl-31829615

Integrin αvß3 and aminopeptidase N (APN, also known as CD13) are two important targets involved in the regulation of angiogenesis, tumor proliferation, invasion, and metastasis. In this study, we developed a heterodimeric tracer consisting of arginine-glycine-aspartic (RGD) and asparagine-glycine-arginine (NGR) peptides targeting αvß3 and CD13, respectively, for PET imaging of breast cancer. The NGR peptide was first modified with N3-NOtB2 and then conjugated to BCN-PEG4-c(RGDyK) via copper-free click chemistry. The resulting precursor was purified and radiolabeled with gallium-68. Small-animal PET/CT imaging and post-imaging biodistribution studies were performed in mice bearing human breast cancer MCF-7, MDA-MB-231, MDA-MB-468, and MX-1 xenografts and pulmonary metastases models. The expression levels of αvß3 and CD13 in tumors were checked via immunochemical staining. The heterodimeric tracer was successfully synthesized and radiolabeled with gallium-68 at a molar activity of 45-100 MBq/nmol at the end of synthesis. It demonstrated high in vitro and in vivo stability. In static PET/CT imaging studies, the MCF-7 tumor could be clearly visualized and exhibited higher uptake at 30 min post injection of 68Ga-NGR-RGD than that of either 68Ga-RGD or 68Ga-NGR alone. High specificity was shown in blocking studies using Arg-Gly-Asp (RGD) and Asp-Gly-Arg (NGR) peptides. The MCF-7 tumor exhibited the highest uptake of 68Ga-NGR-RGD followed by MDA-MB-231, MDA-MB-468, and MX-1 tumors. This was consistent with their expression levels of CD13 and αvß3 as confirmed by western blot and immunohistochemical staining. Metastatic lesions in the lungs were clearly detectable on 68Ga-NGR-RGD PET/CT imaging in mouse models of pulmonary metastases. 68Ga-NGR-RGD, a CD13 and αvß3 dual-receptor targeting tracer, showed higher binding avidities, targeting efficiency, and longer tumor retention time compared with monomeric 68Ga-NGR and 68Ga-RGD. Its promising in vivo performance makes it an ideal candidate for future clinical translation.


Breast Neoplasms/diagnostic imaging , CD13 Antigens/metabolism , Integrin alphaVbeta3/metabolism , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/administration & dosage , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD13 Antigens/antagonists & inhibitors , Cell Line, Tumor , Female , Gallium Radioisotopes , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays
...