Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oncol ; 12: 818437, 2022.
Article in English | MEDLINE | ID: mdl-35530312

ABSTRACT

The lack of inadequate preclinical models remains a limitation for cancer drug development and is a primary contributor to anti-cancer drug failures in clinical trials. Heterotypic multicellular spheroids are three-dimensional (3D) spherical structures generated by self-assembly from aggregates of two or more cell types. Compared to traditional monolayer cell culture models, the organization of cells into a 3D tissue-like structure favors relevant physiological conditions with chemical and physical gradients as well as cell-cell and cell-extracellular matrix (ECM) interactions that recapitulate many of the hallmarks of cancer in situ. Epidermal growth factor receptor (EGFR) mutations are prevalent in non-small cell lung cancer (NSCLC), yet various mechanisms of acquired resistance, including epithelial-to-mesenchymal transition (EMT), limit the clinical benefit of EGFR tyrosine kinase inhibitors (EGFRi). Improved preclinical models that incorporate the complexity induced by epithelial-to-mesenchymal plasticity (EMP) are urgently needed to advance new therapeutics for clinical NSCLC management. This study was designed to provide a thorough characterization of multicellular spheroids of isogenic cancer cells of various phenotypes and demonstrate proof-of-principle for the applicability of the presented spheroid model to evaluate the impact of cancer cell phenotype in drug screening experiments through high-dimensional and spatially resolved imaging mass cytometry (IMC) analyses. First, we developed and characterized 3D homotypic and heterotypic spheroid models comprising EGFRi-sensitive or EGFRi-resistant NSCLC cells. We observed that the degree of EMT correlated with the spheroid generation efficiency in monocultures. In-depth characterization of the multicellular heterotypic spheroids using immunohistochemistry and high-dimensional single-cell analyses by IMC revealed intrinsic differences between epithelial and mesenchymal-like cancer cells with respect to self-sorting, spatiotemporal organization, and stromal cell interactions when co-cultured with fibroblasts. While the carcinoma cells harboring an epithelial phenotype self-organized into a barrier sheet surrounding the fibroblasts, mesenchymal-like carcinoma cells localized to the central hypoxic and collagen-rich areas of the compact heterotypic spheroids. Further, deep-learning-based single-cell segmentation of IMC images and application of dimensionality reduction algorithms allowed a detailed visualization and multiparametric analysis of marker expression across the different cell subsets. We observed a high level of heterogeneity in the expression of EMT markers in both the carcinoma cell populations and the fibroblasts. Our study supports further application of these models in pre-clinical drug testing combined with complementary high-dimensional single-cell analyses, which in turn can advance our understanding of the impact of cancer-stroma interactions and epithelial phenotypic plasticity on innate and acquired therapy resistance in NSCLC.

2.
Tumour Biol ; 35(2): 1365-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24046091

ABSTRACT

The present study examines the effect of the endogenous neuroendocrine factor, corticotropin-releasing factor (CRF), alone or in combination with 5-fluorouracil (5-FU), on 4T1 mammary tumor cells in vitro and in vivo. CRF has been detected in breast cancer tissues; however, the biological effects reported in the literature are sparse and variable. We found that exogenously administered CRF significantly reduced tumor growth without influencing angiogenesis or cell death. Furthermore, CRF reduced tumor interstitial fluid pressure (Pif) and potentiated the effect of 5-FU. These results show that CRF has antitumor effect on mammary carcinoma in mice.


Subject(s)
Breast Neoplasms/drug therapy , Corticotropin-Releasing Hormone/administration & dosage , Fluorouracil/administration & dosage , Mammary Neoplasms, Animal/drug therapy , Animals , Apoptosis , Breast Neoplasms/pathology , Female , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Neovascularization, Pathologic
3.
Target Oncol ; 7(4): 233-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23054400

ABSTRACT

Hypoxia is a critical hallmark of solid tumors and involves enhanced cell survival, angiogenesis, glycolytic metabolism, and metastasis. Hyperbaric oxygen (HBO) treatment has for centuries been used to improve or cure disorders involving hypoxia and ischemia, by enhancing the amount of dissolved oxygen in the plasma and thereby increasing O(2) delivery to the tissue. Studies on HBO and cancer have up to recently focused on whether enhanced oxygen acts as a cancer promoter or not. As oxygen is believed to be required for all the major processes of wound healing, one feared that the effects of HBO would be applicable to cancer tissue as well and promote cancer growth. Furthermore, one also feared that exposing patients who had been treated for cancer, to HBO, would lead to recurrence. Nevertheless, two systematic reviews on HBO and cancer have concluded that the use of HBO in patients with malignancies is considered safe. To supplement the previous reviews, we have summarized the work performed on HBO and cancer in the period 2004-2012. Based on the present as well as previous reviews, there is no evidence indicating that HBO neither acts as a stimulator of tumor growth nor as an enhancer of recurrence. On the other hand, there is evidence that implies that HBO might have tumor-inhibitory effects in certain cancer subtypes, and we thus strongly believe that we need to expand our knowledge on the effect and the mechanisms behind tumor oxygenation.


Subject(s)
Hyperbaric Oxygenation , Neoplasms/therapy , Oxygen/therapeutic use , Animals , Cell Hypoxia , Cell Respiration/drug effects , Clinical Trials as Topic , Humans , Neoplasms/complications
4.
BMC Cancer ; 12: 21, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22251838

ABSTRACT

BACKGROUND: The tumor microenvironment is pivotal in tumor progression. Thus, we aimed to develop a mammary tumor model to elucidate molecular characteristics in the stroma versus the tumor cell compartment by global gene expression. Secondly, since tumor hypoxia influences several aspects of tumor pathophysiology, we hypothesized that hyperoxia might have an inhibitory effect on tumor growth per se. Finally, we aimed to identify differences in gene expression and key molecular mechanisms, both in the native state and following treatment. METHODS: 4T1 dsRed breast cancer cells were injected into eGFP expressing NOD/SCID mice. Group 1 was exposed to 3 intermittent HBO treatments (Day 1, 4 and 7), Group 2 to 7 daily HBO treatments (both 2.5 bar, 100% O2, à 90 min), whereas the controls were exposed to a normal atmosphere. Tumor growth, histology, vascularisation, cell proliferation, cell death and metastasis were assessed. Fluorescence-activated cell sorting was used to separate tumor cells from stromal cells prior to gene expression analysis. RESULTS: The purity of sorted cells was verified by fluorescence microscopy. Gene expression profiling demonstrated that highly expressed genes in the untreated tumor stroma included constituents of the extracellular matrix and matrix metalloproteinases. Tumor growth was significantly inhibited by HBO, and the MAPK pathway was found to be significantly reduced. Immunohistochemistry indicated a significantly reduced microvessel density after intermittent HBO, whereas daily HBO did not show a similar effect. The anti-angiogenic response was reflected in the expression trends of angiogenic factors. CONCLUSIONS: The present in vivo mammary tumor model enabled us to separate tumor and stromal cells, and demonstrated that the two compartments are characterized by distinct gene expressions, both in the native state and following HBO treatments. Furthermore, hyperoxia induced a significant tumor growth-inhibitory effect, with significant down-regulation of the MAPK pathway. An anti-angiogenic effect after intermittent HBO was observed, and reflected in the gene expression profile.


Subject(s)
Cell Hypoxia/physiology , Gene Expression Profiling , Mammary Neoplasms, Experimental/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Extracellular Matrix/genetics , Female , Green Fluorescent Proteins/metabolism , Hyperbaric Oxygenation , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred NOD , Mice, SCID
5.
BMC Cancer ; 9: 446, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-20017908

ABSTRACT

BACKGROUND: Hypoxia is associated with increased resistance to chemo- and radiation-therapy. Hyperoxic treatment (hyperbaric oxygen) has previously been shown to potentiate the effect of some forms of chemotherapy, and this has been ascribed to enhanced cytotoxicity or neovascularisation. The aim of this study was to elucidate whether hyperoxia also enhances any actual uptake of 5FU (5-fluorouracil) into the tumor tissue and if this can be explained by changes in the interstitium and extracellular matrix. METHODS: One group of tumor bearing rats was exposed to repeated hyperbaric oxygen (HBO) treatment (2 bar, pO(2)= 2 bar, 4 exposures à 90 min), whereas one group was exposed to one single identical HBO treatment. Animals housed under normal atmosphere (1 bar, pO(2) = 0.2 bar) served as controls. Three doses of 5FU were tested for dose response. Uptake of [3H]-5FU in the tumor was assessed, with special reference to factors that might have contributed, such as interstitial fluid pressure (Pif), collagen content, oxygen stress (measured as malondialdehyd levels), lymphatics and transcapillary transport in the tumors. RESULTS: The uptake of the cytostatic agent increases immediately after a single HBO treatment (more than 50%), but not 24 hours after the last repeated HBO treatment. Thus, the uptake is most likely related to the transient increase in oxygenation in the tumor tissue. Factors like tumor Pif and collagen content, which decreased significantly in the tumor interstitium after repeated HBO treatment, was without effect on the drug uptake. CONCLUSION: We showed that hyperoxia increases the uptake of [3H]-5FU in DMBA-induced mammary tumors per se, independently of changes in Pif, oxygen stress, collagen fibril density, or transendothelial transport alone. The mechanism by which such an uptake occur is still not elucidated, but it is clearly stimulated by elevated pO(2).


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/physiopathology , Extracellular Fluid/physiology , Fluorouracil/pharmacokinetics , Hyperoxia/physiopathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/physiopathology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , Antimetabolites, Antineoplastic/pharmacokinetics , Blood Volume/physiology , Dose-Response Relationship, Drug , Female , Hyperbaric Oxygenation , Hyperoxia/metabolism , Lymph/physiology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Osmotic Pressure/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Stromal Cells/pathology
6.
Scand J Clin Lab Invest ; 68(7): 602-11, 2008.
Article in English | MEDLINE | ID: mdl-19378432

ABSTRACT

BACKGROUND: Systemic treatment of malignancies with high doses of tumour necrosis factor-alpha (TNFalpha) has an anticancer effect, but also serious side effects. The aim of the present study was to elucidate the effects of local TNFalpha administration alone or in combination with chemotherapy on tumour stroma structure and physiology in di-methyl-benz-anthracene (DMBA)-induced mammary carcinomas in rats. METHODS: TNFalpha (500 ng/mL in a volume of 5 microL) was given s.c. around the carcinoma and 5-fluorouracil (5-FU) (1.5 mg/kg, volume of 0.2 mL) was given i.p on days 1, 4, 7 and 10. RESULTS: Treatment with TNFalpha resulted in a significant reduction of tumour interstitial fluid pressure (TIFP: 75-87%, p < 0.02-0.001), as well as in the number of tumour-infiltrating macrophages, extracellular volume (ECV) and collagen fibril density in carcinoma. In addition, pH was lowered in tumours treated with TNFalpha, suggesting decreased aerobic metabolism. Treatment with TNFalpha, however, had no effect on tumour growth, arterial blood pressure, tumour vessel density, plasma volume or body weight. Concentrations of locally produced VEGF and IL-1beta in carcinoma interstitial fluid or in serum were not affected by TNFalpha. The study demonstrated that these cytokines are produced locally in the tumour. Furthermore, TNFalpha had no effect on efficacy of treatment with 5-FU. CONCLUSIONS: Locally administered TNFalpha did not affect DMBA-induced mammary tumour growth or vasculature, but reduced inflammation and ECM structure, suggesting the latter to be of importance in the observed reduction in TIFP.


Subject(s)
Mammary Neoplasms, Experimental/drug therapy , Tumor Necrosis Factor-alpha/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Collagen/ultrastructure , Extracellular Fluid/chemistry , Female , Interleukin-1beta/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/physiopathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
7.
BMC Cancer ; 7: 23, 2007 Jan 30.
Article in English | MEDLINE | ID: mdl-17263869

ABSTRACT

BACKGROUND: This study investigated the effects of hyperoxic treatment on growth, angiogenesis, apoptosis, general morphology and gene expression in DMBA-induced rat mammary tumors. METHODS: One group of animals was exposed to normobaric hyperoxia (1 bar, pO2 = 1.0 bar) and another group was exposed to hyperbaric hyperoxia (1.5 bar, pO2 = 1.5 bar). A third group was treated with the commonly used chemotherapeutic drug 5- Fluorouracil (5-FU), whereas animals housed under normal atmosphere (1 bar, pO2 = 0.2 bar) served as controls. All treatments were performed on day 1, 4, 7 and 10 for 90 min. Tumor growth was calculated from caliper measurements. Biological effects of the treatment, was determined by assessment of vascular morphology (immunostaining for von Willebrandt factor) and apoptosis (TUNEL staining). Detailed gene expression profiles were obtained and verified by quantitative rtPCR. RESULTS: Tumor growth was significantly reduced (~57-66 %) after hyperoxic treatment compared to control and even more than 5-FU (~36 %). Light microscopic observations of the tumor tissue showed large empty spaces within the tissue after hyperoxic treatment, probably due to loss of glands as indicated by a strong down-regulation of glandular secretory proteins. A significant reduction in mean vascular density (30-50%) was found after hyperoxic treatment. Furthermore, increased apoptosis (18-21%) was found after hyperoxic treatment. CONCLUSION: Thus, by increasing the pO2 in mammary tumor tissue using normobaric and moderate hyperbaric oxygen therapy, a significant retardation in tumor growth is achieved, by loss of glands, reduction in vascular density and enhanced cell death. Hyperbaric oxygen should therefore be further evaluated as a tumor treatment.


Subject(s)
Apoptosis , Hyperbaric Oxygenation , Mammary Neoplasms, Experimental/therapy , Oxygen/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic/therapy , Rats , Rats, Sprague-Dawley
8.
Cancer Lett ; 210(1): 35-40, 2004 Jul 08.
Article in English | MEDLINE | ID: mdl-15172118

ABSTRACT

We tested the hypothesis that hyperbaric oxygen (HBO) alone and with chemotherapy (5-FU) attenuates tumor growth of DMBA-induced tumors in rats. Six series were performed: (1) Controls (air and vehicle 0.9% NaCl i.p.), (2) 5-FU (0.2 mg/kg i.p.), (3) HBO (2 bar for 90 min and vehicle), (4) HBO and 5-FU, (5) HBO (11 days) and air (next 12 days), (6) HBO (23 days). All treatments were applied on days 1, 4, 7, 10 (Series 1-4), as well as on days 14, 17 and 23 (Series 5-6). Tumor diameter increased by 76.7 and 41.2% in untreated controls and in the 5-FU group, respectively, after 10 days. Tumor size fell by 17-24.2% in the HBO groups and by 35.5% when combined with 5-FU (P < 0.05 compared to HBO). HBO treatment reduced the total number of blood vessels in the tumors. After completion of HBO treatment tumor size increased, but statistically insignificant, during the next 12 days.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Fluorouracil/therapeutic use , Hyperbaric Oxygenation , Mammary Neoplasms, Experimental/pathology , Oxygen/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Cell Division/drug effects , Drug Combinations , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , Microcirculation , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...