Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters








Publication year range
1.
Int J Cancer ; 152(12): 2639-2654, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36733230

ABSTRACT

Ablative radiotherapy is a highly efficient treatment modality for patients with metastatic prostate cancer (PCa). However, a subset of patients does not respond. Currently, this subgroup with bad prognosis cannot be identified before disease progression. We hypothesize that markers indicative of radioresistance, stemness and/or bone tropism may have a prognostic potential to identify patients profiting from metastases-directed radiotherapy. Therefore, circulating tumor cells (CTCs) were analyzed in patients with metastatic PCa (n = 24) during radiotherapy with CellSearch, multicolor flow cytometry and imaging cytometry. Analysis of copy-number alteration indicates a polyclonal CTC population that changes after radiotherapy. CTCs were found in 8 out of 24 patients (33.3%) and were associated with a shorter time to biochemical progression after radiotherapy. Whereas the total CTC count dropped after radiotherapy, a chemokine receptor CXCR4-expressing subpopulation representing 28.6% of the total CTC population remained stable up to 3 months. At once, we observed higher chemokine CCL2 plasma concentrations and proinflammatory monocytes. Additional functional analyses demonstrated key roles of CXCR4 and CCL2 for cellular radiosensitivity, tumorigenicity and stem-like potential in vitro and in vivo. Moreover, a high CXCR4 and CCL2 expression was found in bone metastasis biopsies of PCa patients. In summary, panCK+ CXCR4+ CTCs may have a prognostic potential in patients with metastatic PCa treated with metastasis-directed radiotherapy.


Subject(s)
Bone Neoplasms , Neoplastic Cells, Circulating , Prostatic Neoplasms , Male , Humans , Neoplastic Cells, Circulating/pathology , Biomarkers, Tumor , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/pathology , Prognosis , Bone Neoplasms/pathology , Receptors, CXCR4
2.
Drug Deliv ; 24(1): 162-180, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28156178

ABSTRACT

Nanotechnology can offer different solutions for enhancing the therapeutic efficiency of polyphenols, a class of natural products widely explored for a potential applicability for the treatment of different diseases including cancer. While possessing interesting anticancer properties, polyphenols suffer from low stability and unfavorable pharmacokinetics, and thus suitable carriers are required when planning a therapeutic protocol. In the present review, an overview of the different strategies based on polymeric materials is presented, with the aim to highlight the strengths and the weaknesses of each approach and offer a platform of ideas for researchers working in the field.


Subject(s)
Neoplasms/drug therapy , Polymers/chemistry , Polyphenols/administration & dosage , Polyphenols/chemistry , Animals , Drug Carriers/chemistry , Humans , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanotechnology/methods
3.
Cancer Res ; 76(9): 2637-51, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26984757

ABSTRACT

Radiotherapy is a mainstay of curative prostate cancer treatment, but risks of recurrence after treatment remain significant in locally advanced disease. Given that tumor relapse can be attributed to a population of cancer stem cells (CSC) that survives radiotherapy, analysis of this cell population might illuminate tactics to personalize treatment. However, this direction remains challenging given the plastic nature of prostate cancers following treatment. We show here that irradiating prostate cancer cells stimulates a durable upregulation of stem cell markers that epigenetically reprogram these cells. In both tumorigenic and radioresistant cell populations, a phenotypic switch occurred during a course of radiotherapy that was associated with stable genetic and epigenetic changes. Specifically, we found that irradiation triggered histone H3 methylation at the promoter of the CSC marker aldehyde dehydrogenase 1A1 (ALDH1A1), stimulating its gene transcription. Inhibiting this methylation event triggered apoptosis, promoted radiosensitization, and hindered tumorigenicity of radioresistant prostate cancer cells. Overall, our results suggest that epigenetic therapies may restore the cytotoxic effects of irradiation in radioresistant CSC populations. Cancer Res; 76(9); 2637-51. ©2016 AACR.


Subject(s)
Epigenesis, Genetic/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Prostatic Neoplasms/genetics , Radiation Tolerance/genetics , Retinal Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Comparative Genomic Hybridization , DNA Methylation/radiation effects , Flow Cytometry , Heterografts , Histones/genetics , Histones/radiation effects , Humans , Male , Mice , Mice, Nude , Microscopy, Fluorescence , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/radiation effects , Radiotherapy , Retinal Dehydrogenase/radiation effects
4.
Oncotarget ; 6(33): 34494-509, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26460734

ABSTRACT

Despite recent advances in understanding of the molecular pathogenesis and improvement of treatment techniques, locally advanced head and neck squamous cell carcinoma (HNSCC) remains associated with an unfavorable prognosis. Compelling evidence suggests that cancer stem cells (CSC) may cause tumor recurrence if they are not eradicated by current therapies as radiotherapy or radio-chemotherapy. Recent in vitro studies have demonstrated that CSCs may be protected from treatment-induced death by multiple intrinsic and extrinsic mechanisms. Therefore, early determination of CSC abundance in tumor biopsies prior-treatment and development of therapeutics, which specifically target CSCs, are promising strategies to optimize treatment. Here we provide evidence that aldehyde dehydrogenase (ALDH) activity is indicative for radioresistant HNSCC CSCs. Our study suggests that ALDH+ cells comprise a population that maintains its tumorigenic properties in vivo after irradiation and may provide tumor regrowth after therapy. We found that ALDH activity in HNSCC cells can be attributed, at least in part, to the ALDH1A3 isoform and inhibition of the ALDH1A3 expression by small interfering RNA (siRNA) decreases tumor cell radioresistance. The expression dynamic of ALDH1A3 upon irradiation by either induction or selection of the ALDH1A3 positive population correlates to in vivo curability, suggesting that changes in protein expression during radiotherapy are indicative for tumor radioresistance. Our data indicate that ALDH1A3+ HNSCC cells may contribute to tumor relapse after irradiation, and inhibition of this cell population might improve therapeutic response to radiotherapy.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Neoplastic Stem Cells/pathology , Radiation Tolerance/physiology , Animals , Biomarkers, Tumor/analysis , Blotting, Western , Carcinoma, Squamous Cell/enzymology , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , Head and Neck Neoplasms/enzymology , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Squamous Cell Carcinoma of Head and Neck , Tissue Array Analysis , Xenograft Model Antitumor Assays
5.
Int J Cancer ; 137(10): 2492-503, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26059723

ABSTRACT

Many tumors including prostate cancer are maintained by cancer stem cells (CSCs), which might cause tumor relapse if not eradicated during the course of treatment. Specific targeting or radiosensitization of CSCs bear promise to improve tumor curability by synergistic effects in combination with radiotherapy. Carbon nanotubes (CNTs) can be used as promising drug delivery systems for anticancer drugs such as the flavonoid catechin. Catechin is an extensively studied active ingredient of the different plants, including green tea, and it is widely recognized as co-adjuvant in cancer therapy. Here we describe the synthesis of biocompatible, catechin-loaded and gelatin-conjugated CNTs (Gel_CT_CNTs) with anticancer properties and demonstrate their potential for the eradication of prostate CSCs in combination with X-ray irradiation. Gel_CT_CNTs showed a significant enhancement of in vitro anticancer activity as compared to catechin alone. Moreover, treatment of prostate cancer cells with Gel_CT_CNT nanohybrids inhibited the tumorigenic cell population defined by a high aldehyde dehydrogenase (ALDH) activity. A combination of X-ray irradiation and treatment with Gel_CT_CNTs caused a decrease in the protein level of stem cell-related transcription factors and regulators including Nanog, Oct4 and ß-catenin and led to an increase of cancer cell radiosensitivity as demonstrated by clonogenic and spherogenic cell survival assays. Taken together, our results suggest that a combination of irradiation and Gel_CT_CNTs can be potentially used for the radiosensitization and eradication of prostate CSC populations.


Subject(s)
Antineoplastic Agents/chemical synthesis , Catechin/chemistry , Gelatin/chemistry , Nanotubes, Carbon/chemistry , Prostatic Neoplasms/therapy , Radiation-Sensitizing Agents/chemical synthesis , Aldehyde Dehydrogenase/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation , Chemoradiotherapy/methods , Drug Compounding , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Prostatic Neoplasms/pathology , Radiation-Sensitizing Agents/administration & dosage
6.
Cancer Res ; 75(7): 1482-94, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25670168

ABSTRACT

Radiotherapy is a curative treatment option in prostate cancer. Nevertheless, patients with high-risk prostate cancer are prone to relapse. Identification of the predictive biomarkers and molecular mechanisms of radioresistance bears promise to improve cancer therapies. In this study, we show that aldehyde dehydrogenase (ALDH) activity is indicative of radioresistant prostate progenitor cells with an enhanced DNA repair capacity and activation of epithelial-mesenchymal transition (EMT). Gene expression profiling of prostate cancer cells, their radioresistant derivatives, ALDH(+) and ALDH(-) cell populations revealed the mechanisms, which link tumor progenitors to radioresistance, including activation of the WNT/ß-catenin signaling pathway. We found that expression of the ALDH1A1 gene is regulated by the WNT signaling pathway and co-occurs with expression of ß-catenin in prostate tumor specimens. Inhibition of the WNT pathway led to a decrease in ALDH(+) tumor progenitor population and to radiosensitization of cancer cells. Taken together, our results indicate that ALDH(+) cells contribute to tumor radioresistance and their molecular targeting may enhance the effectiveness of radiotherapy.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Neoplastic Stem Cells/enzymology , Prostatic Neoplasms/enzymology , beta Catenin/physiology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/radiation effects , Prostatic Neoplasms/pathology , Radiation Tolerance , Retinal Dehydrogenase , Transcriptome , Wnt Signaling Pathway
7.
Semin Cancer Biol ; 31: 16-27, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24956577

ABSTRACT

Similar to normal tissue, many tumors have a hierarchical organization where tumorigenic cancer stem cells (CSCs) differentiate into non-tumorigenic progenies. A host of studies have demonstrated that although CSCs and their non-tumorigenic progenies within the same clone can share common genotype, they display different epigenetic profiles that results in changes of multiple signaling pathways. Many of these pathways confer cell adaptation to the microenvironmental stresses including inflammation, hypoxia, low pH, shortage in nutrients and anti-cancer therapies. Treatment strategies based on combination of conventional therapies targeting bulk tumor cells and CSC-specific pathway inhibition bear a promise to improve cancer cure compared to monotherapies. In this review we describe the mechanisms of CSC-related therapy resistance including drug efflux by ABC transporters, activation of aldehyde dehydrogenase and developmental pathways, enhanced DNA damage response, autophagy and microenvironmental conditions, and discuss possible therapeutic strategies for improving cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/metabolism , Biomarkers, Tumor/metabolism , Humans , Models, Biological , Molecular Targeted Therapy/methods , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects
8.
Int J Radiat Biol ; 90(8): 687-99, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24650104

ABSTRACT

PURPOSE: Radioresistance of cancer cells remains a fundamental barrier for maximum efficient radiotherapy. Tumor heterogeneity and the existence of distinct cell subpopulations exhibiting different genotypes and biological behaviors raise difficulties to eradicate all tumorigenic cells. Recent evidence indicates that a distinct population of tumor cells, called cancer stem cells (CSC), is involved in tumor initiation and recurrence and is a putative cause of tumor radioresistance. There is an urgent need to identify the intrinsic molecular mechanisms regulating the generation and maintenance of resistance to radiotherapy, especially within the CSC subset. The chemokine C-X-C motif receptor 4 (CXCR4) has been found to be a prognostic marker in various types of cancer, being involved in chemotaxis, stemness and drug resistance. The interaction of CXCR4 with its ligand, the chemokine C-X-C motif ligand 12 (CXCL12), plays an important role in modulating the tumor microenvironment, angiogenesis and CSC niche. Moreover, the therapeutic inhibition of the CXCR4/CXCL12 signaling pathway is sensitizing the malignant cells to conventional anti-cancer therapy. CONTENT: Within this review we are summarizing the role of the CXCR4/CXCL12 axis in the modulation of CSC properties, the regulation of the tumor microenvironment in response to irradiation, therapy resistance and tumor relapse. CONCLUSION: In light of recent findings, the inhibition of the CXCR4/CXCL12 signaling pathway is a promising therapeutic option to refine radiotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Radiation Tolerance , Receptors, CXCR4/metabolism , Animals , Humans , Neoplastic Stem Cells/drug effects , Radiation Tolerance/drug effects , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/radiation effects
9.
Int J Radiat Biol ; 90(8): 659-77, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24524284

ABSTRACT

PURPOSE: Cancer is a multigene disease which arises as a result of mutational and epigenetic changes coupled with activation of complex signaling networks. The use of biomarkers for early cancer detection, staging and individualization of therapy might improve patient care. A few fundamental issues such as tumor heterogeneity, a highly dynamic nature of the intrinsic and extrinsic determinants of radio- and chemoresistance, along with the plasticity and diversity of cancer stem cells (CSC) make biomarker development a challenging task. In this review we outline the preclinical strategies of cancer biomarker discovery including genomic, proteomic, metabolomic and microRNomic profiling, comparative genome hybridization (CGH), single nucleotide polymorphism (SNP) analysis, high throughput screening (HTS) and next generation sequencing (NGS). Other promising approaches such as assessment of circulating tumor cells (CTC), analysis of CSC-specific markers and cell-free circulating tumor DNA (ctDNA) are also discussed. CONCLUSIONS: The emergence of powerful proteomic and genomic technologies in conjunction with advanced bioinformatic tools allows the simultaneous analysis of thousands of biological molecules. These techniques yield the discovery of new tumor signatures, which are sensitive and specific enough for early cancer detection, for monitoring disease progression and for proper treatment selection, paving the way to individualized cancer treatment.


Subject(s)
Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Genetic Techniques , Humans , Mutation , Neoplasms/pathology
10.
Onco Targets Ther ; 6: 1347-61, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24124379

ABSTRACT

The chemokine CXCL12 (SDF-1) and its cell surface receptor CXCR4 were first identified as regulators of lymphocyte trafficking to the bone marrow. Soon after, the CXCL12/CXCR4 axis was proposed to regulate the trafficking of breast cancer cells to sites of metastasis. More recently, it was established that CXCR4 plays a central role in cancer cell proliferation, invasion, and dissemination in the majority of malignant diseases. The stem cell concept of cancer has revolutionized the understanding of tumorigenesis and cancer treatment. A growing body of evidence indicates that a subset of cancer cells, referred to as cancer stem cells (CSCs), plays a critical role in tumor initiation, metastatic colonization, and resistance to therapy. Although the signals generated by the metastatic niche that regulate CSCs are not yet fully understood, accumulating evidence suggests a key role of the CXCL12/CXCR4 axis. In this review we focus on physiological functions of the CXCL12/CXCR4 signaling pathway and its role in cancer and CSCs, and we discuss the potential for targeting this pathway in cancer management.

11.
PLoS One ; 8(5): e65163, 2013.
Article in English | MEDLINE | ID: mdl-23741479

ABSTRACT

Transforming growth factor-ß (TGFß) is a potent regulator of tumorigenesis, although mechanisms defining its tumor suppressing and tumor promoting activities are not understood. Here we describe phosphoproteome profiling of TGFß signaling in mammary epithelial cells, and show that 60 identified TGFß-regulated phosphoproteins form a network with scale-free characteristics. The network highlighted interactions, which may distribute signaling inputs to regulation of cell proliferation, metabolism, differentiation and cell organization. In this report, we identified two novel and TGFß-dependent phosphorylation sites of 14-3-3σ, i.e. Ser69 and Ser74. We observed that 14-3-3σ phosphorylation is a feed-forward mechanism in TGFß/Smad3-dependent transcription. TGFß-dependent 14-3-3σ phosphorylation may provide a scaffold for the formation of the protein complexes which include Smad3 and p53 at the Smad3-specific CAGA element. Furthermore, breast tumor xenograft studies in mice and radiobiological assays showed that phosphorylation of 14-3-3σ at Ser69 and Ser74 is involved in regulation of cancer progenitor population and radioresistance in breast cancer MCF7 cells. Our data suggest that TGFß-dependent phosphorylation of 14-3-3σ orchestrates a functional interaction of TGFß/Smad3 with p53, plays a role in the maintenance of cancer stem cells and could provide a new potential target for intervention in breast cancer.


Subject(s)
14-3-3 Proteins/metabolism , Biomarkers, Tumor/metabolism , Exonucleases/metabolism , Gene Expression Regulation , Radiation Tolerance , Transcription, Genetic , Transforming Growth Factor beta1/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Exoribonucleases , Female , Gene Expression Regulation/drug effects , Heterografts , Humans , MCF-7 Cells , Mice , Models, Biological , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Interaction Maps , Proteome , Signal Transduction/drug effects , Smad3 Protein/metabolism , Transcriptional Activation , Transforming Growth Factor beta1/pharmacology , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL