Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Clin Transl Med ; 12(1): e724, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35090088

RESUMEN

Due to the heterogeneity and high frequency of genome mutations in cancer cells, targeting vital protumour factors found in stromal cells in the tumour microenvironment may represent an ideal strategy in cancer therapy. However, the regulation and mechanisms of potential targetable therapeutic candidates need to be investigated. An in vivo study demonstrated that loss of pentraxin 3 (PTX3) in stromal cells significantly decreased the metastasis and growth of cancer cells. Clinically, our results indicate that stromal PTX3 expression correlates with adverse prognostic features and is associated with worse survival outcomes in triple-negative breast cancer (TNBC). We also found that transforming growth factor beta 1 (TGF-ß1) induces PTX3 expression by activating the transcription factor CCAAT/enhancer binding protein delta (CEBPD) in stromal fibroblasts. Following PTX3 stimulation, CD44, a PTX3 receptor, activates the downstream ERK1/2, AKT and NF-κB pathways to specifically contribute to the metastasis/invasion and stemness of TNBC MDA-MB-231 cells. Two types of PTX3 inhibitors were developed to disrupt the PTX3/CD44 interaction and they showed a significant effect on attenuating growth and restricting the metastasis/invasion of MDA-MB-231 cells, suggesting that targeting the PTX3/CD44 interaction could be a new strategy for future TNBC therapies.


Asunto(s)
Proteína C-Reactiva/efectos de los fármacos , Receptores de Hialuranos/efectos de los fármacos , Componente Amiloide P Sérico/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/genética , Proteína C-Reactiva/genética , Femenino , Humanos , Receptores de Hialuranos/genética , Componente Amiloide P Sérico/genética , Neoplasias de la Mama Triple Negativas/terapia , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
2.
Cell Death Dis ; 11(10): 831, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33024087

RESUMEN

Most luminal breast carcinomas (BrCas) bearing PIK3CA mutations initially respond to phosphoinositide-3-kinase (PI3K)-α inhibitors, but many eventually become resistant. The underlying mechanisms of this resistance remain obscure. In this work, we showed that a CD44high state due to aberrant isoform splicing was acquired from adaptive resistance to a PI3Kα inhibitor (BLY719) in luminal BrCas. Notably, the expression of CD44 was positively correlated with estrogen receptor (ER) activity in PIK3CA-mutant breast cancers, and ER-dependent transcription upon PI3Kα pathway inhibition was in turn mediated by CD44. Furthermore, the interaction of CD44 with the ligand hyaluronan (HA) initiated the Src-ERK signaling cascade, which subsequently maintained AKT and mTOR activity in the presence of a PI3Kα inhibitor. Activation of this pathway was prevented by disruption of the CD44/HA interaction, which in turn restored sensitivity to BLY719. Our results revealed that an ER-CD44-HA signaling circuit that mediates robust compensatory activation of the Src-ERK signaling cascade may contribute to the development of acquired resistance to PI3Kα inhibitors. This study provides new insight into the mechanism of adaptive resistance to PI3Kα inhibition therapy.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Receptores de Hialuranos/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/efectos de los fármacos
3.
Can Respir J ; 2020: 2479369, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849930

RESUMEN

Objective: To investigate the effects of Apatinib on the "stemness" of lung cancer cells in vivo and to explore its related mechanisms. Methods: A xenograft model of lung cancer cells A549 was established in nude mice and randomized into a control group (n = 4) and an Apatinib group (n = 4). Tumor tissues were harvested after 2 weeks, and mRNA was extracted to detect changes in stemness-related genes (CD133, EPCAM, CD13, CD90, ALDH1, CD44, CD45, SOX2, NANOG, and OCT4) and Wnt/ß-catenin, Hedgehog, and Hippo signal pathways. Results: Compared with the control group, the volume and weight of nude mice treated with Apatinib were different and had statistical significance. Apatinib inhibited the expressions of ABCG2, CD24, ICAM-1, OCT4, and SOX2 and upregulated the expressions of CD44, CD13, and FOXD3. Apatinib treatment also inhibited the Wnt/ß-catenin, Hedgehog, and Hippo signaling pathways. Conclusion: Apatinib suppressed the growth of non-small-cell lung cancer cells by repressing the stemness of lung cancer through the inhibition of the Hedgehog, Hippo, and Wnt signaling pathways.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular/efectos de los fármacos , Neoplasias Pulmonares/genética , Células Madre Neoplásicas/efectos de los fármacos , Piridinas/farmacología , Células A549 , Animales , Antígenos CD13/efectos de los fármacos , Antígenos CD13/genética , Antígenos CD13/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Vía de Señalización Hippo , Humanos , Receptores de Hialuranos/efectos de los fármacos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Factores de Transcripción SOXB1/efectos de los fármacos , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Drug Deliv ; 27(1): 128-136, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31894722

RESUMEN

Receptor-mediated active targeting and tumor microenvironment responsive systems from polymeric micelles have been studied for rapid cellular internalization and triggered drug release. Previously we have constructed redox-responsive polymeric micelles composed of vitamin E succinate conjugated hyaluronic acid (HA-ss-TOS), which are able to actively target CD44 proteins and quickly release loaded drugs upon exposure to high levels of glutathione (GSH) in tumor cells. In the present study, we found that despite different cellular internalization mechanisms, micelles showed strong antineoplastic effects on 4T1 and B16F10 cells due to redox responsiveness. HA-ss-TOS-PTX micelles exhibited an excellent tumor targeting ability and prolonged retention time compared to Taxol in vivo. In addition, a superior antitumor effect was achieved compared to PTX-loaded insensitive micelles (HA-TOS-PTX) and Taxol. Our results revealed that PTX-loaded HA-ss-TOS micelles could enhance the antineoplastic efficacy of PTX for breast cancer and melanoma treatment and, thus, deserve further attention.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Portadores de Fármacos/química , Nanopartículas/química , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Línea Celular Tumoral , Supervivencia Celular , Liberación de Fármacos , Humanos , Receptores de Hialuranos/efectos de los fármacos , Ácido Hialurónico/química , Micelas , Oxidación-Reducción , Células Oxífilas/efectos de los fármacos , Tamaño de la Partícula , alfa-Tocoferol/química
5.
Br J Cancer ; 122(3): 361-371, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31772325

RESUMEN

BACKGROUND: ALM201 is a therapeutic peptide derived from FKBPL that has previously undergone preclinical and clinical development for oncology indications and has completed a Phase 1a clinical trial in ovarian cancer patients and other advanced solid tumours. METHODS: In vitro, cancer stem cell (CSC) assays in a range of HGSOC cell lines and patient samples, and in vivo tumour initiation, growth delay and limiting dilution assays, were utilised. Mechanisms were determined by using immunohistochemistry, ELISA, qRT-PCR, RNAseq and western blotting. Endogenous FKBPL protein levels were evaluated using tissue microarrays (TMA). RESULTS: ALM201 reduced CSCs in cell lines and primary samples by inducing differentiation. ALM201 treatment of highly vascularised Kuramochi xenografts resulted in tumour growth delay by disruption of angiogenesis and a ten-fold decrease in the CSC population. In contrast, ALM201 failed to elicit a strong antitumour response in non-vascularised OVCAR3 xenografts, due to high levels of IL-6 and vasculogenic mimicry. High endogenous tumour expression of FKBPL was associated with an increased progression-free interval, supporting the protective role of FKBPL in HGSOC. CONCLUSION: FKBPL-based therapy can (i) dually target angiogenesis and CSCs, (ii) target the CD44/STAT3 pathway in tumours and (iii) is effective in highly vascularised HGSOC tumours with low levels of IL-6.


Asunto(s)
Carcinoma Epitelial de Ovario/patología , Diferenciación Celular/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Neovascularización Patológica/patología , Neoplasias Ováricas/patología , Péptidos/farmacología , Proteínas de Unión a Tacrolimus , Animales , Carcinoma Epitelial de Ovario/irrigación sanguínea , Carcinoma Epitelial de Ovario/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Receptores de Hialuranos/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Técnicas In Vitro , Interleucina-6/metabolismo , Ratones , Ratones SCID , Neovascularización Patológica/metabolismo , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/metabolismo , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Proteínas de Unión a Tacrolimus/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Molecules ; 24(18)2019 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-31509965

RESUMEN

Collagen Tissue Disease-associated Interstitial Lung Fibrosis (CTD-ILDs) and Bronchiolitis Obliterans Syndrome (BOS) represent severe lung fibrogenic disorders, characterized by fibro-proliferation with uncontrolled extracellular matrix deposition. Hyaluronic acid (HA) plays a key role in fibrosis with its specific receptor, CD44, overexpressed by CTD-ILD and BOS cells. The aim is to use HA-liposomes to develop an inhalatory treatment for these diseases. Liposomes with HA of two molecular weights were prepared and characterized. Targeting efficiency was assessed toward CTD-ILD and BOS cells by flow cytometry and confocal microscopy and immune modulation by RT-PCR and ELISA techniques. HA-liposomes were internalized by CTD-ILD and BOS cells expressing CD44, and this effect increased with higher HA MW. In THP-1 cells, HA-liposomes decreased pro-inflammatory cytokines IL-1ß, IL-12, and anti-fibrotic VEGF transcripts but increased TGF-ß mRNA. However, upon analyzing TGF-ß release from healthy donors-derived monocytes, we found liposomes did not alter the release of active pro-fibrotic cytokine. All liposomes induced mild activation of neutrophils regardless of the presence of HA. HA liposomes could be also applied for lung fibrotic diseases, being endowed with low pro-inflammatory activity, and results confirmed that higher MW HA are associated to an increased targeting efficiency for CD44 expressing LFs-derived from BOS and CTD-ILD patients.


Asunto(s)
Bronquiolitis Obliterante/tratamiento farmacológico , Ácido Hialurónico/farmacología , Liposomas/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Células A549 , Adulto , Bronquiolitis Obliterante/patología , Sistemas de Liberación de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Voluntarios Sanos , Humanos , Receptores de Hialuranos/efectos de los fármacos , Ácido Hialurónico/química , Liposomas/química , Microscopía Confocal , Monocitos/efectos de los fármacos , Fibrosis Pulmonar/patología , Factor de Crecimiento Transformador beta/genética , Factor A de Crecimiento Endotelial Vascular/genética
7.
Life Sci ; 196: 56-62, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29355544

RESUMEN

AIMS: Cancer stem cells (CSCs) are considered the prime source of cancer recurrence, metastasis, and progression and represent important targets for developing novel anticancer agents and therapeutic strategies. The aim of this study was to investigate the effect of treating breast CSCs with the anticancer flavonoid, quercetin. MAIN METHODS: We examined changes in the cluster of differentiation CD44+/CD24-CSC population and behavior using the breast cancer cell line MCF-7. KEY FINDINGS: Our results indicated that cell viability, clone formation, mammosphere generation, and nude mice tumor metastasis were inhibited in the CD44+/CD24- population and that MCF-7 cells exhibited G1-phase arrest after quercetin treatment. Additionally, CyclinD1 and B cell lymphoma-2 expression were suppressed and Bcl-2-like protein-4 expression was enhanced after quercetin treatment. We also observed that estrogen receptor α and phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling were downregulated concurrently with the inhibition of CD44+/CD24- viability and clone formation. Our findings suggested that quercetin treatment promoted weaker malignant activity associated with CSCs relative to that observed in normal cancer cells through its inhibition of the PI3K/Akt/mTOR-signaling pathway. SIGNIFICANCE: These results indicated that CSCs are potential therapeutic targets for quercetin treatment of breast cancer.


Asunto(s)
Antioxidantes/farmacología , Antígeno CD24/efectos de los fármacos , Receptores de Hialuranos/efectos de los fármacos , Proteína Oncogénica v-akt/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Quercetina/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Femenino , Fase G1/efectos de los fármacos , Humanos , Células MCF-7 , Ratones Desnudos , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Biomed Pharmacother ; 88: 210-217, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28107698

RESUMEN

Prostate cancer (PCa) is the second most common type of cancer and the fifth leading cause of cancer-related death among men. Development of chemoresistance, tumor relapse and metastasis remain major barriers to effective treatment and all been identified to be associated with cancer stem cells (CSCs). Natural flavonoids such as apigenin have been shown to have the ability to improve the therapeutic efficacy of common chemotherapy agents through CSCs sensitization. Thus, the aim of this study was to evaluate the combination of apigenin with cisplatin on CD44+ PCa stem cell growth and migration. Platinum-based anti-neoplastic drugs have been used to treat a number of malignancies including PCa. However, acquired resistance and side effects unfortunately have limited cisplatin's use. A CD44+ subpopulation was isolated from human androgen-independent PC3 PCa cells by using human CD44-PE antibody. IC50 values were determined by MTT test. RT-qPCR, Western blot analyses and image-based cytometer were used to investigate apoptosis, cell cycle and their underlying molecular mechanisms. Cell migration was evaluated by wound healing test. The combination of the IC50 doses of apigenin (15µM) and cisplatin (7.5µM) for 48h significantly enhanced cisplatin's cytotoxic and apoptotic effects through downregulation of Bcl-2, sharpin and survivin; and upregulation of caspase-8, Apaf-1 and p53 mRNA expression. The combined therapy suppressed the phosphorylation of p-PI3K and p-Akt, inhibited the protein expression of NF-κB, and downregulated the cell cycle by upregulating p21, as well as cyclin dependent kinases CDK-2, -4, and -6. Apigenin significantly increased the inhibitory effects of cisplatin on cell migration via downregulation of Snail expression. In conclusion, our study showed the possible therapeutic approach of using apigenin to potentially increase the effects of cisplatin by targeting CSCs subset in prostate cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Apigenina/farmacología , Cisplatino/uso terapéutico , Receptores de Hialuranos/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Receptores de Hialuranos/efectos de los fármacos , Masculino , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/patología , Cicatrización de Heridas/efectos de los fármacos
9.
Arch Oral Biol ; 74: 69-74, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27886571

RESUMEN

OBJECTIVE: Cancer stem cells contribute to tumor recurrence, and a hypoxic environment is critical for maintaining cancer stem cells. Apigenin is a natural product with anticancer activity. However, the effect of apigenin on cancer stem cells remains unclear. Our aim was to investigate the effect of apigenin on cancer stem cell marker expression in head and neck squamous cell carcinoma cells under hypoxia. DESIGN: We used three head and neck squamous cell carcinoma cell lines; HN-8, HN-30, and HSC-3. The mRNA expression of cancer stem cell markers was determined by semiquantitative RT-PCR and Real-time PCR. The cytotoxic effect of apigenin was determined by MTT colorimetric assay. Flow cytometry was used to reveal the number of cells expressing cancer stem cell surface markers. RESULTS: HN-30 cells, a cancer cell line from the pharynx, showed the greatest response to hypoxia by increasing their expression of CD44, CD105, NANOG, OCT-4, REX-1, and VEGF. Apigenin significantly decreased HN-30 cell viability in dose- and time-dependent manners. In addition, 40µM apigenin significantly down-regulated the mRNA expression of CD44, NANOG, and CD105. Consistent with these results, the hypoxia-induced increase in CD44+ cells, CD105+ cells, and STRO-1+ cells was significantly abolished by apigenin. CONCLUSION: Apigenin suppresses cancer stem cell marker expression and the number of cells expressing cell surface markers under hypoxia.


Asunto(s)
Apigenina/farmacología , Biomarcadores/metabolismo , Carcinoma de Células Escamosas/metabolismo , Hipoxia de la Célula/genética , Neoplasias de Cabeza y Cuello/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Endoglina/efectos de los fármacos , Endoglina/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Humanos , Receptores de Hialuranos/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Factores de Transcripción de Tipo Kruppel/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteína Homeótica Nanog/efectos de los fármacos , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Faringe , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Medicine (Baltimore) ; 95(43): e5150, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27787370

RESUMEN

BACKGROUND: Flavopiridol a semisynthetic flavone that inhibits cyclin-dependent kinases (CDKs) and has growth-inhibitory activity and induces a blockade of cell-cycle progression at G1-phase and apoptosis in numerous human tumor cell lines and is currently under investigation in phase II clinical trials. Cancer stem cells (CSCs) are comprised of subpopulation of cells in tumors that have been proposed to be responsible for recurrence and resistance to chemotherapy. The aim of the present study was to investigate the effects of flavopiridol in cancer stem cell cytoskeleton, cell adhesion, and epithelial to mesenchymal transition in CSCs. METHODS: The cells were treated with flavopiridol to determine the inhibitory effect. Cell viability and proliferation were determined by using the WST-1 assay. Caspase activity and immunofluorescence analyses were performed for the evaluation of apoptosis, cell cytoskeleton, and epithelial-mesenchymal transition (EMT) markers. The effects of flavopiridol on the cell cycle were also evaluated. Flow cytometric analysis was used to detect the percentages of CSCs subpopulation. We analyzed the gene expression patterns to predict cell cycle and cell cytoskeleton in CSCs by RT-PCR. RESULTS: Flavopiridol-induced cytotoxicity and apoptosis at the IC50 dose, resulting in a significant increase expression of caspases activity. Cell cycle analyses revealed that flavopiridol induces G1 phase cell cycle arrest. Flavopiridol significantly decreased the mRNA expressions of the genes that regulate the cell cytoskeleton and cell cycle components and cell motility in CSCs. CONCLUSION: Our results suggest that Flavopiridol has activity against lung CSCs and may be effective chemotherapeutic molecule for lung cancer treatment.


Asunto(s)
Antígeno AC133/metabolismo , Carcinoma de Células Escamosas/patología , Flavonoides/farmacología , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/efectos de los fármacos , Piperidinas/farmacología , Antígeno AC133/efectos de los fármacos , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Caspasas/biosíntesis , Caspasas/genética , Línea Celular Tumoral , Proliferación Celular , ADN de Neoplasias/genética , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Hialuranos/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Reacción en Cadena de la Polimerasa
11.
J Oral Pathol Med ; 45(3): 180-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26399460

RESUMEN

BACKGROUND: CD44 and aldehyde dehydrogenase 1 (ALDH1) have been shown to be useful markers for identification of cancer stem cells (CSCs). We previously reported that glycogen synthase kinase 3ß (GSK3ß) is involved in regulation of the self-renewal ability of head and neck squamous cell carcinoma (HNSCC) CSCs. The purpose of the present study was to clarify the role of GSK3ß in CD44(high) /ALDH1(high) HNSCC cells. METHODS: Cells with greater expression of CD44 and higher ALDH1 enzymatic activity were FACS sorted from the OM-1 HNSCC cell line. The self-renewal ability of CD44(high) /ALDH1(high) cells was then examined using a tumor sphere formation assay. mRNA expressions of the stem cell markers Sox2, Oct4, and Nanog, as well as GSK3ß were evaluated by real-time RT-PCR. RESULTS: CD44(high) /ALDH1(high) cells exhibited higher tumor sphere forming ability and increased expression of stem cell markers as compared with CD44(high) /ALDH1(low) cells. Interestingly, spindle-shaped cells positive for vimentin were found in the CD44(high) /ALDH1(high) but not the CD44(high) /ALDH1(low) cell population. In addition, the ALDH1 activity and sphere forming ability of CD44(high) /ALDH1(high) cells was significantly inhibited by GSK3ß knockdown. On the other hand, CD44(high) /ALDH1(low) cells exhibited high epidermal growth factor receptor (EGFR) expression and increased cell growth. CONCLUSIONS: Our results show that GSK3ß plays a major role in maintenance of stemness of CD44(high) /ALDH1(high) HNSCC cells. Additionally, they indicate a close relationship between CSC and mesenchymal characteristics in HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Receptores de Hialuranos/biosíntesis , Isoenzimas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Retinal-Deshidrogenasa/efectos de los fármacos , Familia de Aldehído Deshidrogenasa 1 , Biomarcadores de Tumor/biosíntesis , Carcinoma de Células Escamosas/enzimología , Línea Celular Tumoral , Activación Enzimática , Receptores ErbB/biosíntesis , Neoplasias de Cabeza y Cuello/enzimología , Humanos , Receptores de Hialuranos/efectos de los fármacos , Isoenzimas/biosíntesis , Isoenzimas/metabolismo , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/metabolismo , Proteína Homeótica Nanog/biosíntesis , Células Madre Neoplásicas/enzimología , Factores de Transcripción de Octámeros/biosíntesis , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/genética , Retinal-Deshidrogenasa/biosíntesis , Retinal-Deshidrogenasa/metabolismo , Factores de Transcripción SOXB2/biosíntesis , Carcinoma de Células Escamosas de Cabeza y Cuello
12.
Curr Drug Deliv ; 13(4): 500-11, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26549038

RESUMEN

BACKGROUND: Hyaluronan (HA), a glycosaminoglycan, is a key extracellular matrix (ECM) component, and has been established to contribute to fibrotic, angiogenic, inflammatory as well as processes supporting cancer development. The changes in HA deposition in different tumors have been widely studied. Indeed, a multitude of reports demonstrate that HA expression is increased in different neoplasmatic tissues including lung, colon, prostate and breast cancer. The aims of this paper are to critically and in depth discuss aspects of HA metabolism in cancer and recent developments of its utilization in cancer therapy. METHODS: Up to date research and online content are reviewed. RESULTS: The cellular roles of HA are perpetrated through molecular interactions with HA-binding proteins, called hyaladherins, including CD44 receptor as well as receptor for hyaluronan-mediated motility (RHAMM). HA binding can be followed by receptor-mediated endocytosis. Importantly, hyaladherins show an altered expression in tumor tissues. Indeed, post-translational alterations in CD44 structure have been suggested to regulate the equilibrium between the "inactive" low affinity state and the "active" high affinity state of the HA binding capacity. In this concept HA fragments can be utilized as specific targeting ligands for efficient and safe drug delivery in cancer. CONCLUSION: HA-drug bioconjugates and nanoparticles have emerged as a promising platform for drug delivery during cancer treatment as demonstrated in various pre-clinical studies. Recent developments from clinical trials indicate that the utilization of specific HA-drug bioconjugates might be approved for the medical practice in the nearest future.


Asunto(s)
Antineoplásicos/uso terapéutico , Portadores de Fármacos , Ácido Hialurónico/administración & dosificación , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Proteínas de la Matriz Extracelular/efectos de los fármacos , Humanos , Receptores de Hialuranos/efectos de los fármacos
13.
Laryngoscope ; 126(5): E184-90, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26690734

RESUMEN

OBJECTIVES/HYPOTHESIS: To evaluate the efficacy of peritumoral hyaluronic acid (HA)-cisplatin therapy in a murine model of laryngeal squamous cell carcinoma and to evaluate its effect on cancer stem cells (CSCs). STUDY DESIGN: An orthotopic murine study utilizing University of Michigan squamous cell carcinoma-12 (UMSCC-12) laryngeal cancer cells was conducted in randomized controlled fashion with three treatment arms: saline, systemic cisplatin, and peritumoral HA-cisplatin. METHODS: UMSCC-12 laryngeal cancer cells were inoculated into the buccal mucosa of athymic nude mice followed by weekly treatment with saline, systemic cisplatin, or peritumoral HA-cisplatin for 3 weeks. Tumor response and animal weight was monitored and change in CD44 proportion was analyzed ex vivo. RESULTS: HA-cisplatin demonstrated superior antitumor efficacy and greater reduction in CD44 positivity on ex vivo analysis. CONCLUSIONS: Peritumoral nanoconjugated HA-cisplatin provides superior antitumor efficacy compared to standard cisplatin therapy in an in vivo laryngeal cancer model. There was also selective targeting of CD44+ cancer cells with HA-cisplatin. This therapeutic strategy could represent the first selective laryngeal CSC-targeted therapy. Further preclinical investigation is warranted to evaluate its role for locally advanced head and neck cancer treatment. LEVEL OF EVIDENCE: NA Laryngoscope, 126:E184-E190, 2016.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Cisplatino/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias Laríngeas/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Animales , Humanos , Receptores de Hialuranos/efectos de los fármacos , Ácido Hialurónico/farmacología , Ratones , Ratones Desnudos , Nanoconjugados/administración & dosificación , Carcinoma de Células Escamosas de Cabeza y Cuello
14.
Integr Biol (Camb) ; 7(12): 1547-60, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26456171

RESUMEN

Fragments of the extracellular matrix component hyaluronan (HA) promote tissue inflammation, fibrosis and tumor progression. HA fragments act through HA receptors including CD44, LYVE1, TLR2, 4 and the receptor for hyaluronan mediated motility (RHAMM/HMMR). RHAMM is a multifunctional protein with both intracellular and extracellular roles in cell motility and proliferation. Extracellular RHAMM binds directly to HA fragments while intracellular RHAMM binds directly to ERK1 and tubulin. Both HA and regions of tubulin (s-tubulin) are anionic and bind to basic amino acid-rich regions in partner proteins, such as in HA and tubulin binding regions of RHAMM. We used this as a rationale for developing bioinformatics and SPR (surface plasmon resonance) based screening to identify high affinity anionic RHAMM peptide ligands. A library of 12-mer peptides was prepared based on the carboxyl terminal tail sequence of s-tubulin isoforms and assayed for their ability to bind to the HA/tubulin binding region of recombinant RHAMM using SPR. This approach resulted in the isolation of three 12-mer peptides with nanomolar affinity for RHAMM. These peptides bound selectively to RHAMM but not to CD44 or TLR2,4 and blocked RHAMM:HA interactions. Furthermore, fluorescein-peptide uptake by PC3MLN4 prostate cancer cells was blocked by RHAMM mAb but not by CD44 mAb. These peptides also reduced the ability of prostate cancer cells to degrade collagen type I. The selectivity of these novel HA peptide mimics for RHAMM suggest their potential for development as HA mimetic imaging and therapeutic agents for HA-promoted disease.


Asunto(s)
Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Movimiento Celular/fisiología , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Secuencia de Aminoácidos , Materiales Biomiméticos/farmacología , Neoplasias de la Mama/metabolismo , Carbocianinas , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Femenino , Colorantes Fluorescentes , Humanos , Receptores de Hialuranos/efectos de los fármacos , Ligandos , Masculino , Datos de Secuencia Molecular , Invasividad Neoplásica/prevención & control , Biblioteca de Péptidos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Resonancia por Plasmón de Superficie , Tubulina (Proteína)/farmacología
15.
Ginekol Pol ; 86(5): 388-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26117979

RESUMEN

Ovarian carcinoma constitutes the main cause of cancer-related death among women. The curability rates remain low despite rapid advances in medicine. Thus, the search for new and improved methods continues, with CD44-targeting as one of them. CD44 is a cell-surface glycoprotein, which binds to its ligand--hyaluronic acid (HA)--and regulates crucial processes such as cell differentiation, proliferation and migration. Overexpression of CD44, observed in many ovarian cancer cells, is used in creating carriers for selective delivery of various drugs (paclitaxel, doxorubicin, camptothecin or cisplatin) to cancer cells. In this article, we summarized the current state of knowledge regarding CD44-targeting as a new and more efficient way of ovarian cancer treatment, with high potential and promising therapeutic perspectives.


Asunto(s)
Antineoplásicos/uso terapéutico , Receptores de Hialuranos/efectos de los fármacos , Ácido Hialurónico/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Terapias en Investigación , Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Neoplasias Ováricas/metabolismo
16.
Glycoconj J ; 32(7): 549-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25997408

RESUMEN

Hyaluronan (HA) is an integral component of the extracellular matrix. Its interactions with a cell surface receptor CD44 has been shown to play important roles in a variety of biological events including cell proliferation and metastasis. As multivalent CD44-HA binding is critical for downstream signaling, compounds that can selectively disrupt the complex formation of HA polysaccharide with CD44 can serve as useful probes of CD44 mediated cellular events as well as potential leads for novel therapeutics. Herein, we report the synthesis of several series of HA conjugates to target the HA binding pocket of CD44. As a small library of HA disaccharide derivatives failed to exhibit any inhibitory activities, we focused on HA tetrasaccharide based analogs. Traditional synthetic strategies towards HA oligosaccharides involve the construction of backbone from the corresponding monosaccharide building blocks, which can be quite tedious. In order to expedite the synthesis, we designed a new synthetic route taking advantage of the ability of hyaluronidase to generate large quantities of HA tetrasaccharide through digestion of HA polysaccharides. The HA tetrasaccharide obtained was utilized to prepare multiple S-linked HA analogs bearing aromatic groups at the reducing end glycan. One such compound containing an m-benzyl phenyl moiety exhibited significant inhibition of CD44-HA binding. Our approach provides a new direction towards the design of HA based CD44 antagonists.


Asunto(s)
Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Línea Celular Tumoral , Proliferación Celular , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Humanos , Receptores de Hialuranos/química , Receptores de Hialuranos/efectos de los fármacos , Ácido Hialurónico/síntesis química , Ácido Hialurónico/genética , Hialuronoglucosaminidasa/química , Hialuronoglucosaminidasa/metabolismo , Metástasis de la Neoplasia , Oligosacáridos/química , Oligosacáridos/genética , Transducción de Señal
17.
Biomaterials ; 53: 763-74, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25890771

RESUMEN

Multi-drug delivery focuses on different signaling pathways in cancer cells that have synergistic anti-proliferative effects. In this study, we developed multi-prodrug nanocarriers (MPDNCs) consisting of poly (l-lysine)-carboxylate PTX (PLL-PTX) and hyaluronic acid-conjugated GEM (HA-GEM) for CD44-targeted synergistic biliary cancer therapy. An in vitro study of cell viability and mRNA expression levels and an in vivo study showed that MPDNCs more effectively inhibit proliferation in CD44-overexpressing cancer cells (HuCCT1) than in cells with lower CD44 expression (SCK) by synergistically inducing apoptosis. Consequently, these results demonstrate that MPDNCs are prodrugs with synergistic cancer therapeutic efficacy and effective cellular uptake at target cells compared to free drugs, indicating their strong potential as efficient multi-drug-carrying nano-platforms for cancer treatment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias del Sistema Biliar/tratamiento farmacológico , Portadores de Fármacos , Receptores de Hialuranos/efectos de los fármacos , Nanopartículas , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Sistema Biliar/patología , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Humanos , Paclitaxel/administración & dosificación , Gemcitabina
18.
Int J Biol Macromol ; 72: 510-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25224289

RESUMEN

The main aim of this work was to evaluate a nanoconjugate system of paclitaxel loaded self-assembling, biodegradable micelles for targeting CD44 overexpression in cancer cells. The shape and size, zeta potential, encapsulation efficiency and cell uptake of these drug-loaded micelles were evaluated. To understand their bio distribution profile, the hyaluronate (HA) micelles were labeled with Flamma™-774 NIR dye and injected into SCC7 tumor induced mice. Cell viability in response to drug loaded and unloaded micelles was studied in SCC7 cancer cells using the MTS assay. An in vivo tumor inhibition study was conducted by intravenous injection of paclitaxel-loaded HA micelle nanoparticles as well as control nanoparticles without paclitaxel. The shape of the nanomicelles was evaluated by loading them with hydrophobic superparamagnetic iron oxide nanoparticle and then visualizing them by TEM. In conclusion, paclitaxel-loaded HA nanoparticulate micelles might be found to be a specific and efficient chemotherapeutic treatment for CD44 overexpressing cancer cells.


Asunto(s)
Terapia Molecular Dirigida , Nanopartículas/administración & dosificación , Neoplasias/tratamiento farmacológico , Paclitaxel/administración & dosificación , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Humanos , Receptores de Hialuranos/efectos de los fármacos , Ácido Hialurónico/administración & dosificación , Ácido Hialurónico/química , Ratones , Micelas , Nanopartículas/química , Neoplasias/patología , Paclitaxel/química
19.
Int J Biol Macromol ; 72: 569-74, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25220787

RESUMEN

In present investigation, we developed a novel hyaluronated cationic nanostructured lipid carrier (CNLCs) which contains CPT-11/irinotecan to target CD44 biomarker which commonly overexpresses on colon cancer. The cationic core NLCs was developed by using capmule MCM and compritol as mix lipid phase with 150 ± 2.5 nm particles size and 93.98 ± 2.5% entrapment efficiency. The cationic core NLCs were coated with hyaluronic acid by ionic conjugation. The particle size of hyaluronated CNLCs was enhanced to 255 ± 4.2 nm. In-vitro drug release studies revealed a slow and sustain release of the drug and a practically no leaching of coumarin-6. The confocal laser microscopy studies were performed with developed CNLCs in two CD44 overexpressing cell lines (HT-29 and Colo-320). The studies revealed marked uptake of the CNLCs by both the cell lines in just two hours. The results were comparable to the FITC conjugated CD44 antibodies. Therefore, the developed dual purpose CNLCs were found to be highly specific for CD44 biomarker. The developed formulations were found to be compatible with blood components. The MTT assay revealed that the developed particles had enhanced cytotoxicity against non-MDR as well MDR cancer cells.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Ácido Hialurónico/administración & dosificación , Adenocarcinoma/genética , Adenocarcinoma/patología , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Camptotecina/química , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Cumarinas/administración & dosificación , Cumarinas/química , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Humanos , Receptores de Hialuranos/efectos de los fármacos , Ácido Hialurónico/química , Irinotecán , Lípidos/administración & dosificación , Lípidos/química , Nanopartículas/administración & dosificación , Nanopartículas/química , Tiazoles/administración & dosificación , Tiazoles/química
20.
Expert Opin Ther Pat ; 24(7): 821-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24798704

RESUMEN

INTRODUCTION: The CD44 transmembrane glycoprotein family mediates cellular responses to the microenvironment through binding of hyaluronic acid (HA) and other proteins of the extracellular matrix. These interactions start intracellular signaling cascades that foster tumor growth, survival and spread. AREAS COVERED: The patent concerns the use of a humanized anti-CD44 mAb (RG7356) to treat patients with aggressive forms of chronic lymphocytic leukemia (CLL). The RG7356 humanized antibody was designed to bind the constant region of CD44, preventing binding with HA. The interruption of this circuit in vitro is followed by the induction of caspase-dependent apoptosis in leukemic cells. In agreement with a functional association between CD44 and zeta-associated protein of 70 kDa (ZAP-70) in activating intracellular signaling, the strongest effects were observed in ZAP-70(+) CLL cells, generally associated to a poor prognosis. Furthermore, these effects were confirmed using in vivo models, where CLL cells were xenografted in immunocompromised mice. EXPERT OPINION: In summary, these studies suggest that this new humanized mAb may provide additional clinical benefit to CLL patients receiving current standard treatments, specifically to those with a more aggressive disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Receptores de Hialuranos/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Humanos , Receptores de Hialuranos/inmunología , Patentes como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA