Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J R Soc Interface ; 19(190): 20220088, 2022 05.
Article in English | MEDLINE | ID: mdl-35612280

ABSTRACT

Epidermal growth factor (EGF) signalling regulates normal epithelial and other cell growth, with EGF receptor (EGFR) overexpression reported in many cancers. However, the role of EGFR clusters in cancer and their dependence on EGF binding is unclear. We present novel single-molecule total internal reflection fluorescence microscopy of (i) EGF and EGFR in living cancer cells, (ii) the action of anti-cancer drugs that separately target EGFR and human EGFR2 (HER2) on these cells and (iii) EGFR-HER2 interactions. We selected human epithelial SW620 carcinoma cells for their low level of native EGFR expression, for stable transfection with fluorescent protein labelled EGFR, and imaged these using single-molecule localization microscopy to quantify receptor architectures and dynamics upon EGF binding. Prior to EGF binding, we observe pre-formed EGFR clusters. Unexpectedly, clusters likely contain both EGFR and HER2, consistent with co-diffusion of EGFR and HER2 observed in a different model CHO-K1 cell line, whose stoichiometry increases following EGF binding. We observe a mean EGFR : EGF stoichiometry of approximately 4 : 1 for plasma membrane-colocalized EGFR-EGF that we can explain using novel time-dependent kinetics modelling, indicating preferential ligand binding to monomers. Our results may inform future cancer drug developments.


Subject(s)
Epidermal Growth Factor , ErbB Receptors , Carcinoma/metabolism , Cell Line, Tumor , Colonic Neoplasms/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Humans , Phosphorylation , Receptor, ErbB-2/metabolism , Signal Transduction
2.
Anticancer Res ; 40(11): 6063-6073, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33109544

ABSTRACT

BACKGROUND/AIM: Interactions between colorectal cancer (CRC) cells and myofibroblasts govern many processes such as cell growth, migration, invasion and differentiation, and contribute to CRC progression. Robust experimental tests are needed to investigate the nature of these interactions for future anticancer studies. The purpose of the study was to design and validate in vitro assays for studying the communication between myofibroblasts and CRC epithelial cell lines. MATERIALS AND METHODS: The influence of co-culture of myofibroblasts and CRC cell lines is discussed using various in vitro assays including direct co-culture, transwell assays, Matrigel-based differentiation and cell invasion experiments. RESULTS: The results from these in vitro assays clearly demonstrated various aspects of the crosstalk between myofibroblasts and CRC cell lines, which include cell growth, differentiation, migration and invasion. CONCLUSION: The reported in vitro assays provide a basis for investigating the factors that control the myofibroblast-epithelial cell interactions in CRC in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Communication/drug effects , Colon/pathology , Colorectal Neoplasms/pathology , Myofibroblasts/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Humans , Male , Neoplasm Invasiveness
3.
Front Biosci (Landmark Ed) ; 25(9): 1765-1786, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32472757

ABSTRACT

Placental alkaline phosphatase, PLAP encoded by ALPP gene in humans is mainly expressed in placenta and testis, and not expressed in any other normal tissues. PLAP is overexpressed in colorectal cancers which makes it an attractive target for CAR (chimeric antigen receptor)-T cell therapy. PLAP mRNA expression was detected in 21.5% (25 out of 116) of colorectal cancer cell lines and this expression was confirmed by FACS at the protein level. In addition, IHC staining on primary colorectal cancer tumors demonstrated PLAP expression in >20% of colorectal cancer tumors. We generated mouse and humanized PLAP ScFv-CAR-T cells and demonstrated high specificity against PLAP-positive colon cancer cells using RTCA (real-time cytotoxicity assay) and IFN-gamma secretion. In addition, humanized-CAR-T cells significantly decreased Lovo xenograft tumor growth in vivo. The combination of hPLAP-CAR-T cells with PD-1, PD-L1 or LAG-3 checkpoint inhibitors significantly increased the activity of hPLAP-CAR-T cells. This study demonstrates ability of novel PLAP-CAR-T cells to kill colorectal cancers and that the extent of killing can be increased by combination with checkpoint inhibitors.


Subject(s)
Alkaline Phosphatase/immunology , Colonic Neoplasms/immunology , Isoenzymes/immunology , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/immunology , T-Lymphocytes/immunology , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/metabolism , Animals , Caco-2 Cells , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/therapy , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy, Adoptive/methods , Interferon-gamma/immunology , Interferon-gamma/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Chimeric Antigen/metabolism , Single-Chain Antibodies/metabolism , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays/methods
4.
Cancer Biol Med ; 16(2): 234-246, 2019 May.
Article in English | MEDLINE | ID: mdl-31516745

ABSTRACT

OBJECTIVE: Patients with BRAF-mutant colorectal cancer (CRC) have a poor prognosis. Molecular status is not currently used to select which drug to use in combination with radiotherapy. Our aim was to identify drugs that radiosensitise CRC cells with known BRAF status. METHODS: We screened 298 oncological drugs with and without ionising radiation in colorectal cancer cells isogenic for BRAF. Hits from rank product analysis were validated in a 16-cell line panel of human CRC cell lines, using clonogenic survival assays and xenograft models in vivo. RESULTS: Most consistently identified hits were drugs targeting cell growth/proliferation or DNA damage repair. The most effective class of drugs that radiosensitised wild-type and mutant cell lines was PARP inhibitors. In clonogenic survival assays, talazoparib produced a radiation enhancement ratio of 1.9 in DLD1 (BRAF-wildtype) cells and 1.8 in RKO (BRAF V600E) cells. In DLD1 xenografts, talazoparib significantly increased the inhibitory effect of radiation on tumour growth (P ≤ 0.01). CONCLUSIONS: Our method for screening large drug libraries for radiosensitisation has identified PARP inhibitors as promising radiosensitisers of colorectal cancer cells with wild-type and mutant BRAF backgrounds.

5.
J Pathol ; 247(3): 293-304, 2019 03.
Article in English | MEDLINE | ID: mdl-30306567

ABSTRACT

Colonic epithelial cells are highly polarised with a lumen-facing apical membrane, termed the brush border, and a basal membrane in contact with the underlying extracellular matrix (ECM). This polarity is often maintained in cancer tissue in the form of neoplastic glands and has prognostic value. We compared the cellular polarity of several ex vivo spheroid colonic cancer cultures with their parental tumours and found that those grown as non-attached colonies exhibited apical brush border proteins on their outer cellular membranes. Transfer of these cultures to an ECM, such as collagen, re-established the centralised apical polarity observed in vivo. The multidrug resistance protein ABCB1 also became aberrantly polarised to outer colony membranes in suspension cultures, unlike cultures grown in collagen, where it was polarised to central lumens. This polarity switch was dependent on the presence of serum or selected serum components, including epidermal growth factor (EGF), transforming growth factor-ß1 (TGF-ß1) and insulin-like growth factor-1 (IGF-1). The apical/basal orientation of primary cancer colon cultures cultured in collagen/serum was modulated by α2ß1 integrin signalling. The polarisation of ABCB1 in colonies significantly altered drug uptake and sensitivity, as the outward polarisation of ABCB1 in suspension colonies effluxed substrates more effectively than ECM-grown colonies with ABCB1 polarised to central lumens. Thus, serum-free suspension colonies were more resistant to a variety of anti-cancer drugs than ECM-grown colonies. In conclusion, the local stroma, or absence thereof, can have profound effects on the sensitivity of colorectal cultures to drugs that are ABCB1 substrates. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Polarity/physiology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , ATP Binding Cassette Transporter, Subfamily B/physiology , Actins/metabolism , Collagen/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Combinations , Extracellular Matrix/metabolism , Humans , Integrin beta1/physiology , Laminin , Proteoglycans , Signal Transduction/physiology , Spheroids, Cellular/pathology , Tumor Cells, Cultured
6.
Stem Cell Res ; 27: 109-120, 2018 03.
Article in English | MEDLINE | ID: mdl-29414601

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) liver metastasis is highly unfavorable for patient outcome and is a leading cause of cancer-related death. Pre-clinical research of CRC liver metastasis predominately utilizes CRC cell lines grown in tissue culture. Here, we demonstrate that CRC liver metastases organoids derived from human specimens recapitulate some aspects of human disease. METHODS: Human CRC liver metastases pathological specimens were obtained following patient consent. Tumor disaggregates were plated and organoids were allowed to expand. CRC markers were identified by immunofluorescence. Stem cell genes were analysed by QPCR and flow cytometry. Response to drug therapy was quantified using time-lapse imaging and MATLAB analysis. RESULTS: Organoids showed global expression of the epithelial marker, EpCAM and the adenocarcinoma marker, CEA CAM1. Flow cytometry analysis demonstrated that organoids express the stem cell surface markers CD24 and CD44. Finally, we demonstrated that CRC liver metastases organoids acquire chemotherapy resistance and can be utilized as surrogates for drug testing. CONCLUSION: These data demonstrate that CRC liver metastases organoids recapitulate some aspects of human disease and may provide an invaluable resource for investigating novel drug therapies, chemotherapy resistance and mechanism of metastasis.


Subject(s)
Colorectal Neoplasms/complications , Liver Neoplasms/secondary , Organoids/pathology , Cell Proliferation/genetics , Cell Proliferation/physiology , Cells, Cultured , Flow Cytometry , Humans , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
7.
Oncotarget ; 8(63): 106764-106777, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29290987

ABSTRACT

Mitochondria form a highly dynamic network driven by opposing scission and fusion events. DRP1 is an essential modulator of mitochondrial fission and dynamics within mammalian cells. Its fission activity is regulated by posttranslational modifications such as activating phosphorylation at serine 616. DRP1 activity has recently been implicated as being dysregulated in numerous human disorders such as cancer and neurodegenerative diseases. Here we describe the development of a cell-based screening assay to detect DRP1 activation. We utilized this to undertake focused compound library screening and identified potent modulators that affected DRP1 activity including ICG-001, which is described as WNT/ß-catenin signaling inhibitor. Our findings elucidate novel details about ICG-001's mechanism of action (MOA) in mediating anti-proliferative activity. We show ICG-001 both inhibits mitochondrial fission and activates an early endoplasmic reticulum (ER) stress response to induce cell death in susceptible colorectal cancer cell lines.

8.
Proc Natl Acad Sci U S A ; 113(15): E2162-71, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27036009

ABSTRACT

Pericryptal myofibroblasts in the colon and rectum play an important role in regulating the normal colorectal stem cell niche and facilitating tumor progression. Myofibroblasts previously have been distinguished from normal fibroblasts mostly by the expression of α smooth muscle actin (αSMA). We now have identified AOC3 (amine oxidase, copper containing 3), a surface monoamine oxidase, as a new marker of myofibroblasts by showing that it is the target protein of the myofibroblast-reacting mAb PR2D3. The normal and tumor tissue distribution and the cell line reactivity of AOC3 match that expected for myofibroblasts. We have shown that the surface expression of AOC3 is sensitive to digestion by trypsin and collagenase and that anti-AOC3 antibodies can be used for FACS sorting of myofibroblasts obtained by nonenzymatic procedures. Whole-genome microarray mRNA-expression profiles of myofibroblasts and skin fibroblasts revealed four additional genes that are significantly differentially expressed in these two cell types: NKX2-3 and LRRC17 in myofibroblasts and SHOX2 and TBX5 in skin fibroblasts. TGFß substantially down-regulated AOC3 expression in myofibroblasts but in skin fibroblasts it dramatically increased the expression of αSMA. A knockdown of NKX2-3 in myofibroblasts caused a decrease of myofibroblast-related gene expression and increased expression of the fibroblast-associated gene SHOX2, suggesting that NKX2-3 is a key mediator for maintaining myofibroblast characteristics. Our results show that colorectal myofibroblasts, as defined by the expression of AOC3, NKX2-3, and other markers, are a distinctly different cell type from TGFß-activated fibroblasts.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Cell Adhesion Molecules/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Amine Oxidase (Copper-Containing)/genetics , Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Colon/cytology , Colon/metabolism , Colorectal Neoplasms/metabolism , Fibroblasts/cytology , Gene Expression , Homeodomain Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Oligonucleotide Array Sequence Analysis , Proteins/genetics , RNA, Messenger/metabolism , Skin/cytology , Skin/metabolism , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
9.
Clin Cancer Res ; 22(13): 3286-97, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26861458

ABSTRACT

PURPOSE: CEA TCB is a novel IgG-based T-cell bispecific (TCB) antibody for the treatment of CEA-expressing solid tumors currently in phase I clinical trials (NCT02324257). Its format incorporates bivalent binding to CEA, a head-to-tail fusion of CEA- and CD3e-binding Fab domains and an engineered Fc region with completely abolished binding to FcγRs and C1q. The study provides novel mechanistic insights into the activity and mode of action of CEA TCB. EXPERIMENTAL DESIGN: CEA TCB activity was characterized on 110 cell lines in vitro and in xenograft tumor models in vivo using NOG mice engrafted with human peripheral blood mononuclear cells. RESULTS: Simultaneous binding of CEA TCB to tumor and T cells leads to formation of immunologic synapses, T-cell activation, secretion of cytotoxic granules, and tumor cell lysis. CEA TCB activity strongly correlates with CEA expression, with higher potency observed in highly CEA-expressing tumor cells and a threshold of approximately 10,000 CEA-binding sites/cell, which allows distinguishing between high- and low-CEA-expressing tumor and primary epithelial cells, respectively. Genetic factors do not affect CEA TCB activity confirming that CEA expression level is the strongest predictor of CEA TCB activity. In vivo, CEA TCB induces regression of CEA-expressing xenograft tumors with variable amounts of immune cell infiltrate, leads to increased frequency of activated T cells, and converts PD-L1 negative into PD-L1-positive tumors. CONCLUSIONS: CEA TCB is a novel generation TCB displaying potent antitumor activity; it is efficacious in poorly infiltrated tumors where it increases T-cell infiltration and generates a highly inflamed tumor microenvironment. Clin Cancer Res; 22(13); 3286-97. ©2016 AACR.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoembryonic Antigen/immunology , Immunotherapy/methods , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antineoplastic Agents/immunology , Binding Sites/immunology , CD3 Complex/immunology , Cell Line, Tumor , Female , Humans , Lymphocyte Activation/immunology , Mice , Receptors, Fc/immunology , Xenograft Model Antitumor Assays
10.
Oncotarget ; 6(21): 18518-33, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26041882

ABSTRACT

Most solid tumors contain a subfraction of cells with stem/progenitor cell features. Stem cells are naturally chemoresistant suggesting that chronic chemotherapeutic stress may select for cells with increased "stemness". We carried out a comprehensive molecular and functional analysis of six independently selected colorectal cancer (CRC) cell lines with acquired resistance to three different chemotherapeutic agents derived from two distinct parental cell lines. Chronic drug exposure resulted in complex alterations of stem cell markers that could be classified into three categories: 1) one cell line, HT-29/5-FU, showed increased "stemness" and WNT-signaling, 2) three cell lines showed decreased expression of stem cell markers, decreased aldehyde dehydrogenase activity, attenuated WNT-signaling and lost the capacity to form colonospheres and 3) two cell lines displayed prominent expression of ABC transporters with a heterogeneous response for stem cell markers. While WNT-signaling could be attenuated in the HT-29/5-FU cells by the WNT-signaling inhibitors ICG-001 and PKF-118, this was not accompanied by any selective growth inhibitory effect suggesting that the cytotoxic activity of these compounds is not directly linked to WNT-signaling inhibition. We conclude that classical WNT-signaling inhibitors have toxic off-target activities that need to be addressed for clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplastic Stem Cells/drug effects , Wnt Signaling Pathway/drug effects , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Irinotecan , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organoplatinum Compounds/pharmacology , Oxaliplatin , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyrimidinones/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Triazines/pharmacology , Wnt Signaling Pathway/genetics
11.
Cell Cycle ; 13(17): 2697-706, 2014.
Article in English | MEDLINE | ID: mdl-25486357

ABSTRACT

Enzastaurin is a selective inhibitor of protein kinase C ß and a potent inhibitor of tumor angiogenesis. In addition, enzastaurin shows direct cytotoxic activity toward a subset of tumor cells including colorectal cancer cells (CRC). In spite of promising results in animal models, the clinical activity of enzastaurin in CRC patients has been disappointing although a subset of patients seems to derive benefit. In the present study we investigated the biological and cytotoxic activities of enzastaurin toward a panel of well-characterized CRC cell lines in order to clarify the mechanistic basis for the cytotoxic activity. Our results show that enzastaurin is significantly more cytotoxic toward CRC cells with chromosome instability (CIN) compared to cells with microsatellite instability (MSI). Since CIN is usually attributed to mitotic dysfunction, the influence of enzastaurin on cell cycle progression and mitotic transit was characterized for representative CIN and MSI cell lines. Enzastaurin exposure was accompanied by prolonged metaphase arrest in CIN cells followed by the appearance of tetraploid and micronuclei-containing cells as well as by increased apoptosis, whereas no detectable mitotic dysfunctions were observed in MSI cells exposed to isotoxic doses of enzastaurin. Our study identifies enzastaurin as a new, context dependent member of a heterogeneous group of anticancer compounds that induce "mitotic catastrophe," that is mitotic dysfunction accompanied by cell death. These data provide novel insight into the mechanism of action of enzastaurin and may allow the identification of biomarkers useful to identify CRC patients particularly likely, or not, to benefit from treatment with enzastaurin.


Subject(s)
Chromosomal Instability/drug effects , Chromosome Segregation/drug effects , Colorectal Neoplasms/pathology , Indoles/pharmacology , Mitosis/drug effects , Protein Kinase C beta/antagonists & inhibitors , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Centrosome/drug effects , Centrosome/metabolism , Humans , Metaphase/drug effects , Polyploidy , Protein Kinase C beta/metabolism , Tumor Suppressor Protein p53/metabolism
12.
Cancer Res ; 74(20): 5866-77, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25168481

ABSTRACT

Drugs that inhibit insulin-like growth factor 1 (IGFI) receptor IGFIR were encouraging in early trials, but predictive biomarkers were lacking and the drugs provided insufficient benefit in unselected patients. In this study, we used genetic screening and downstream validation to identify the WNT pathway element DVL3 as a mediator of resistance to IGFIR inhibition. Sensitivity to IGFIR inhibition was enhanced specifically in vitro and in vivo by genetic or pharmacologic blockade of DVL3. In breast and prostate cancer cells, sensitization tracked with enhanced MEK-ERK activation and relied upon MEK activity and DVL3 expression. Mechanistic investigations showed that DVL3 is present in an adaptor complex that links IGFIR to RAS, which includes Shc, growth factor receptor-bound-2 (Grb2), son-of-sevenless (SOS), and the tumor suppressor DAB2. Dual DVL and DAB2 blockade synergized in activating ERKs and sensitizing cells to IGFIR inhibition, suggesting a nonredundant role for DVL3 in the Shc-Grb2-SOS complex. Clinically, tumors that responded to IGFIR inhibition contained relatively lower levels of DVL3 protein than resistant tumors, and DVL3 levels in tumors correlated inversely with progression-free survival in patients treated with IGFIR antibodies. Because IGFIR does not contain activating mutations analogous to EGFR variants associated with response to EGFR inhibitors, we suggest that IGF signaling achieves an equivalent integration at the postreceptor level through adaptor protein complexes, influencing cellular dependence on the IGF axis and identifying a patient population with potential to benefit from IGFIR inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Drug Resistance, Neoplasm , Insulin-Like Growth Factor I/physiology , Phosphoproteins/physiology , Receptor, IGF Type 1/antagonists & inhibitors , ras Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Dishevelled Proteins , Gene Expression , Head and Neck Neoplasms/metabolism , Humans , Inhibitory Concentration 50 , Isoxazoles/pharmacology , MAP Kinase Signaling System , Male , Mice , Pyrimidines/pharmacology , Receptor, IGF Type 1/metabolism , Wnt Proteins/metabolism , Xenograft Model Antitumor Assays
13.
J Pathol ; 234(1): 34-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24797403

ABSTRACT

We have developed a simple procedure for deriving pure cultures of growing cancer cells from colorectal cancers, including material refrigerated overnight, for pathological characterization and cytotoxicity assays. Forty-six cancers were processed and cultures set up under varying culture conditions. Use of a Rho kinase (ROCK1) inhibitor markedly increased culture survival, resulting in 80% of samples growing in culture for at least 1 month and beyond. Overnight refrigeration of samples before culture initiation had little effect on success rates, paving the way for cultures to be established for samples collected over wide geographical areas, such as those for clinical trials. Primary cultures demonstrated good correlation for differentiation markers compared to parent cancers, and were highly dynamic in 3D culture. In Matrigel, many colonies formed central lumens, indicating the presence of stem-like cells. Viable colonies in these cultures recapitulated the in vivo generation of carcinoembryonic antigen (CEA)-positive necrotic/apoptotic debris, much of which was derived from abnormal vacuolated dynamic 'bubble cells' that have not previously been described. Although bubble cells morphologically resembled signet ring cells, a rare cancer subtype, immunostaining suggested that they were most likely derived from terminally differentiated enterocytes. Micro-assays showed that drug toxicity could be measured in these cultures within hours and with sensitivity down to a few hundred cells. Primary cultures derived by our method provide valid in vitro avatars for studying the pathology of cancers in vitro and are amenable to pre-clinical drug testing, paving the way for personalized cancer treatment.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Goblet Cells/pathology , Spheroids, Cellular/pathology , Cell Differentiation , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Goblet Cells/drug effects , Humans , Keratin-20/metabolism , Spheroids, Cellular/drug effects , Staurosporine/pharmacology , Time Factors , Tumor Cells, Cultured
14.
Int J Oncol ; 42(5): 1644-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23546019

ABSTRACT

Irinotecan is a major anticancer agent specifically targeting DNA topoisomerase I. Its cytotoxicity is mediated via a two-step process involving accumulation of reversible DNA­topoisomerase I complexes associated with transient DNA single-strand breaks which subsequently are converted into permanent DNA double-strand breaks by the replication fork during S phase. Irinotecan may be selectively active for treatment of colorectal cancers that show microsatellite instability (MSI) due to deficiencies in mismatch repair enzymes, compared to tumors that are microsatellite stable but show chromosome instability (CIN). Although the clinical activity of irinotecan is principally limited by acquired drug resistance, surprisingly little is known about the influence of prolonged irinotecan exposure on the cell cycle dynamics. We have developed two colon cancer cell lines resistant to SN-38, the active metabolite of irinotecan, one derived from HT-29 (CIN), the other from HCT-116 (MSI). We here show that besides classical resistance mechanisms, SN-38 resistance is accompanied by an increased generation doubling time, a decreased S phase fraction and an increased G2 fraction in vitro as in tumor xenografts for both CIN and MSI models. As a consequence, SN-38-resistant cells and tumors show cross-resistance to the S-phase selective agent 5-fluorouracil. The resistance is accompanied by increased basal levels of γ-H2AX and phospho-Chk2 without notable changes in the levels of phospho-Chk1. Taken together, our results show that prolonged irinotecan exposure is accompanied by stable modifications of cell cycle dynamics which could have profound impact on tumor sensitivity to a wide range of antitumor agents and may influence tumor progression in patients.


Subject(s)
Camptothecin/analogs & derivatives , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type I/genetics , HCT116 Cells , Humans , Irinotecan , Microsatellite Instability/drug effects , S Phase/drug effects , S Phase/genetics
15.
Clin Cancer Res ; 17(20): 6522-30, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21880790

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) and VEGF(R) signaling show extensive cross-talk, providing a rationale for joint targeting of the two pathways. However, combinations of monoclonal antibodies (mAb) targeting EGFR and VEGF showed disappointing activity in patients with colorectal cancer (CRC). We speculated that inhibition of surface receptors and ligands might only partly prevent oncogenic signaling whereas small-molecule tyrosine kinase inhibitors (TKI) would also influence intracellular signaling. EXPERIMENTAL DESIGN: Mice with CRC xenografts were treated with two TKIs, vargatef and afatinib, or with two mAbs, bevacizumab and cetuximab, and their influence on tumor growth, viability, in vivo DNA synthesis, and the presence of phosphorylated EGFR and VEGFR was determined. The activity of the TKIs was further characterized in CRC cells with different KRAS status. RESULTS: Vargatef and afatinib together showed strong tumor growth inhibition toward HT-29 xenografts compared with either drug alone, which was associated with a 5-fold increase in apoptotic tumor cell death. In comparison, bevacizumab and cetuximab together were exclusively cytostatic with no more activity than either drug alone. Exposure to the two TKIs was accompanied by a marked decrease of tumor-associated intracellular phospho-VEGFR1 and phospho-EGFR, whereas similar exposure to the two mAbs had no detectable effect. A synergistic activity of vargatef plus afatinib was observed in all eight CRC cell lines examined, independent of KRAS status. CONCLUSIONS: Our results indicate that attenuation of intracellular EGFR and/or VEGF signaling is required for cytotoxic activity. These findings provide a rationale for trials of the TKIs, even in patients with mutant KRAS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Afatinib , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Bevacizumab , Cell Proliferation/drug effects , Cell Survival/drug effects , Cetuximab , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Female , HT29 Cells , Humans , Indoles/pharmacology , Mice , Mice, Nude , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
16.
Mol Cancer Ther ; 10(8): 1481-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21622731

ABSTRACT

PM01183 is a novel marine-derived covalent DNA binder in clinical development. PM01183 is structurally similar to trabectedin (yondelis, ecteinascidin-743) except for the C subunit, and this modification is accompanied by different pharmacokinetics in cancer patients. We here characterize the interaction of PM01183 with the nucleotide excision repair (NER) pathway in comparison with trabectedin. Our results show for the first time that although neither PM01183 nor trabectedin is repaired by NER, both compounds are able to interfere with the NER machinery thereby attenuating the repair of specific NER substrates. We further show that the NER activity is increased in 3 of 4 cellular models with acquired resistance to cisplatin or oxaliplatin, confirming the involvement of NER in the resistance to platinum derivatives. Importantly, both PM01183 and trabectedin show unchanged or even enhanced activity toward all 4 cisplatin- and oxaliplatin-resistant cell lines. We finally show that combinations of PM01183 and cisplatin were mostly synergistic toward both parental and cisplatin-resistant ovarian carcinoma cells as indicated by Chou and Talalay analysis. These data show that the C subunit of trabectedin can be subjected to at least some structural modifications without loss of activity or NER interaction. While PM01183 and trabectedin appear functionally similar in cellular models, it is likely that the differences in pharmacokinetics may allow different dosing and scheduling of PM01183 in the clinic that could lead to novel and/or increased antitumor activity. Taken together, our results provide a mechanistic basis to support clinical trials of PM01183 alone or in combination with cisplatin.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Carbolines/pharmacology , Cisplatin/pharmacology , DNA Repair/drug effects , Dioxoles/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Tetrahydroisoquinolines/pharmacology , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Carbolines/chemistry , Cell Line, Tumor , Chromatin/metabolism , DNA Damage/drug effects , DNA Damage/radiation effects , Dioxoles/chemistry , Dioxoles/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Synergism , HCT116 Cells , HT29 Cells , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Neoplasms/metabolism , Neoplasms/pathology , Organoplatinum Compounds/pharmacology , Oxaliplatin , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/metabolism , Trabectedin , Ultraviolet Rays/adverse effects
17.
Pharmacol Ther ; 131(1): 80-90, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21439312

ABSTRACT

The last decade has witnessed the approval of monoclonal antibodies (mAbs) and small molecule tyrosine kinase inhibitors (TKIs) for targeting of oncogenic signaling pathways. Generally, the clinical activity of these agents has been less than expected, in part due to unsuspected feed-back loops and cross-talk between different signaling pathways, thereby suggesting the interest of inhibiting multiple pathways. The extensive degree of EGFR-VEGF(R) pathway cross-talk identifies these pathways as particularly promising for joint targeting. Activation of the EGFR pathway increases the production of tumor-derived VEGF that acts on endothelial cells in a paracrine manner to promote angiogenesis. Accordingly, exposure to EGFR inhibitors is accompanied by attenuation of VEGF expression while resistance to EGFR inhibitors is frequently associated with enhanced VEGF levels. Recent data have expanded the biological activities of the two pathways by documenting a role for VEGF signaling in tumor cell survival and demonstrating the expression of EGFR by some tumor-associated endothelial cells. At least part of these signaling events are intracrine (intracellular and autocrine) and thus not readily accessible for the mAbs which target extracellular ligands and membrane receptors. This may explain why two major clinical trials combining EGFR and VEGF-targeted mAbs gave disappointing results and suggest a need for compounds that are able to inhibit intracrine signaling. Clinical application of new combinations should be preceded by preclinical development guided by functional biomarker analysis to identify active drug combinations and to facilitate the identification of patient subgroups likely, or not, to respond to dual pathway inhibition.


Subject(s)
Green Fluorescent Proteins/metabolism , Neoplasms/blood supply , Neoplasms/drug therapy , Vascular Endothelial Growth Factors/metabolism , Animals , Cell Survival/drug effects , Green Fluorescent Proteins/genetics , Humans , Molecular Targeted Therapy/methods , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Receptor Cross-Talk , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...