Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cancers (Basel) ; 14(24)2022 Dec 17.
Article in English | MEDLINE | ID: mdl-36551714

ABSTRACT

Mutations in the isocitrate dehydrogenase 1 (IDH1MUT) gene occur in various types of malignancies, including ~60% of chondrosarcomas, ~30% of intrahepatic cholangiocarcinomas and >80% of low-grade gliomas. IDH1MUT are causal in the development and progression of these types of cancer due to neomorphic production of the oncometabolite D-2-hydroxyglutarate (D-2HG). Intracellular accumulation of D-2HG has been implicated in suppressing homologous recombination and renders IDH1MUT cancer cells sensitive to DNA-repair-inhibiting agents, such as poly-(adenosine 5'-diphosphate−ribose) polymerase inhibitors (PARPi). Hyperthermia increases the efficacy of DNA-damaging therapies such as radiotherapy and platinum-based chemotherapy, mainly by inhibition of DNA repair. In the current study, we investigated the additional effects of hyperthermia (42 °C for 1 h) in the treatment of IDH1MUT HCT116 colon cancer cells and hyperthermia1080 chondrosarcoma cancer cells in combination with radiation, cisplatin and/or a PARPi on clonogenic cell survival, cell cycle distribution and the induction and repair of DNA double-strand breaks. We found that hyperthermia in combination with radiation or cisplatin induces an increase in double-strand breaks and cell death, up to 10-fold in IDH1MUT cancer cells compared to IDH1 wild-type cells. This vulnerability was abolished by the IDH1MUT inhibitor AGI-5198 and was further increased by the PARPi. In conclusion, our study shows that IDH1MUT cancer cells are sensitized to hyperthermia in combination with irradiation or cisplatin and a PARPi. Therefore, hyperthermia may be an efficacious sensitizer to cytotoxic therapies in tumors where the clinical application of hyperthermia is feasible, such as IDH1MUT chondrosarcoma of the extremities.

2.
Ther Adv Med Oncol ; 14: 17588359221109196, 2022.
Article in English | MEDLINE | ID: mdl-35782751

ABSTRACT

Purpose: Regorafenib monotherapy, a multikinase inhibitor of angiogenesis, tumor microenvironment, and tumorigenesis, showed promising results in gastric cancer. We aimed to assess the tolerability of regorafenib and paclitaxel in patients with advanced esophagogastric cancer (EGC) refractory to first-line treatment, and explore potential biomarkers. Methods: Patients received paclitaxel (80 mg/m2) on days 1, 8, and 15 of a 28-day cycle and regorafenib (80/120/160 mg) on days 1-21 in the dose-escalation cohort, and the maximum-tolerated dose (MTD) in the dose-expansion cohort. Exploratory, overall survival (OS) and progression-free survival (PFS) were compared to a propensity-score matched cohort receiving standard second-/third-line systemic treatment. Paclitaxel pharmacokinetics were assessed using samples from day 1 (D1) and day 15 (D15). We performed enzyme-linked immunosorbent assay measurements of galectin-1, RNA sequencing, and shallow whole-genome sequencing of metastatic tumor biopsies for biomarker analyses. Results: In the dose-escalation cohort (n = 14), the MTD of regorafenib was 120 mg. In all, 34 patients were enrolled in the dose-expansion cohort. Most common toxicities (all grades; grade ⩾ 3) were fatigue (79%; 4%) and sensory neuropathy (63%; 4%). Best responses achieved were partial response (28%) and stable disease (54%). Median OS and PFS were 7.8 and 4.2 months, respectively (median follow-up: 7.8 months). OS (p = 0.08) and PFS (p = 0.81) were not significantly improved compared to the matched cohort. Paclitaxel concentrations were significantly increased with regorafenib (D15) compared with paclitaxel only (D1; p < 0.05); no associations were observed with toxicity or efficacy. An increase in circulating galectin-1 compared to baseline was associated with shorter OS (p < 0.01). Enrichment of angiogenesis-related gene expression was observed in short survivors measured by RNA sequencing. Chromosome 19q13.12-q13.2 amplification was associated with shorter OS (p = 0.02) and PFS (p = 0.02). Conclusion: Treatment with regorafenib and paclitaxel is tolerable and shows promising efficacy in advanced EGC refractory to first-line treatment. Galectin-1 and chromosome 19q13.12-q13.2 amplification could serve as negative predictive biomarkers for treatment response. Registration: Clinicaltrials.gov, NCT02406170, https://clinicaltrials.gov/ct2/show/NCT02406170.

3.
Cancers (Basel) ; 13(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069550

ABSTRACT

BACKGROUND: Mutations in isocitrate dehydrogenase 1 (IDH1) occur in 60% of chondrosarcoma, 80% of WHO grade II-IV glioma and 20% of intrahepatic cholangiocarcinoma. These solid IDH1-mutated tumors produce the oncometabolite D-2-hydroxyglutarate (D-2HG) and are more vulnerable to disruption of their metabolism. METHODS: Patients with IDH1-mutated chondrosarcoma, glioma and intrahepatic cholangiocarcinoma received oral combinational treatment with the antidiabetic drug metformin and the antimalarial drug chloroquine. The primary objective was to determine the occurrence of dose-limiting toxicities (DLTs) and the maximum tolerated dose (MTD). Radiological and biochemical tumor responses to metformin and chloroquine were investigated using CT/MRI scans and magnetic resonance spectroscopy (MRS) measurements of D-2HG levels in serum. RESULTS: Seventeen patients received study treatment for a median duration of 43 days (range: 7-74 days). Of twelve evaluable patients, 10 patients discontinued study medication because of progressive disease and two patients due to toxicity. None of the patients experienced a DLT. The MTD was determined to be 1500 mg of metformin two times a day and 200 mg of chloroquine once a day. A serum D/L-2HG ratio of ≥4.5 predicted the presence of an IDH1 mutation with a sensitivity of 90% and a specificity of 100%. By utilization of digital droplet PCR on plasma samples, we were able to detect tumor-specific IDH1 hotspot mutations in circulating tumor DNA (ctDNA) in investigated patients. CONCLUSION: Treatment of advanced IDH1-mutated solid tumors with metformin and chloroquine was well tolerated but did not induce a clinical response in this phase Ib clinical trial.

4.
J Histochem Cytochem ; 68(1): 33-57, 2020 01.
Article in English | MEDLINE | ID: mdl-31566074

ABSTRACT

Glioblastoma is the most aggressive primary brain tumor. Slowly dividing and therapy-resistant glioblastoma stem cells (GSCs) reside in protective peri-arteriolar niches and are held responsible for glioblastoma recurrence. Recently, we showed similarities between GSC niches and hematopoietic stem cell (HSC) niches in bone marrow. Acute myeloid leukemia (AML) cells hijack HSC niches and are transformed into therapy-resistant leukemic stem cells (LSCs). Current clinical trials are focussed on removal of LSCs out of HSC niches to differentiate and to become sensitized to chemotherapy. In the present study, we elaborated further on these similarities by immunohistochemical analyses of 17 biomarkers in paraffin sections of human glioblastoma and human bone marrow. We found all 17 biomarkers to be expressed both in hypoxic peri-arteriolar HSC niches in bone marrow and hypoxic peri-arteriolar GSC niches in glioblastoma. Our findings implicate that GSC niches are being formed in glioblastoma as a copy of HSC niches in bone marrow. These similarities between HSC niches and GSC niches provide a theoretic basis for the development of novel strategies to force GSCs out of their niches, in a similar manner as in AML, to induce GSC differentiation and proliferation to render them more sensitive to anti-glioblastoma therapies.


Subject(s)
Bone Marrow Cells/cytology , Glioblastoma/immunology , Glioblastoma/pathology , Stem Cell Niche , Animals , Cell Line, Tumor , Glioblastoma/therapy , Hematopoietic Stem Cells/pathology , Humans , Optical Imaging , Tumor Hypoxia
5.
Biotechniques ; 67(4): 172-176, 2019 10.
Article in English | MEDLINE | ID: mdl-31560238

ABSTRACT

In biomedical research, large-scale profiling of gene expression has become routine and offers a valuable means to evaluate changes in onset and progression of diseases, in particular cancer. An overwhelming amount of cancer genomics data has become publicly available, and the complexity of these data makes it a challenge to perform in silico data exploration, integration and analysis, in particular for scientists lacking a background in computational programming or informatics. Many web interface tools make these large datasets accessible but are limited to process large datasets. To accelerate the translation of genomic data into new insights, we provide a simple method to explore and select data from cancer genomic datasets to generate gene-expression profiles of subsets that are of specific genetic, biological or clinical interest.


Subject(s)
Genomics/methods , Neoplasms/genetics , Transcriptome , DNA Methylation , Data Visualization , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Internet , Isocitrate Dehydrogenase/genetics , L-Lactate Dehydrogenase/genetics , Mutation , Neoplasms/metabolism , Software
6.
Acta Histochem ; 121(2): 125-134, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30454859

ABSTRACT

Immunohistochemistry (IHC) specifically localizes proteins in cells and tissues, but methodologies vary widely. Therefore, we performed a methodological IHC optimization and validation study. First, we compared advantages and disadvantages of cryostat sections versus paraffin sections. Second, we compared and optimized antigen retrieval in paraffin sections using citrate buffer and Tris/EDTA buffer. Third, aminoethyl carbazole (AEC) and 3,3'-diaminobenzidine (DAB) were tested as horseradish peroxidase (HRP) substrates to obtain a water-insoluble coloured end product to visualize antigens. Fourth, secondary antibodies conjugated with either mono-HRP or poly-HRP were compared. The study was performed using serial sections of human tonsil. IHC was performed with primary antibodies against endothelial cell marker CD31, smooth muscle actin (SMA), chemokine stromal-derived factor-1α (SDF-1α) and its receptor C-X-C receptor type 4 (CXCR4), macrophage marker CD68 and proliferation marker Ki67. DAB rather than AEC, and cryostat sections rather than paraffin sections gave optimum staining at highest primary antibody dilutions, whereas tissue morphology in paraffin sections was superior. Loss of antigenicity in paraffin sections by formaldehyde fixation, heat and/or masking of epitopes was counteracted by antigen retrieval but not for all antigens. Two out of six antigens (CD31 and CD68) could not be retrieved irrespective time and type of retrieval. Tris-EDTA was superior to citrate buffer for antigen retrieval. The use of mono-HRP or poly-HRP depended on the affinity of the primary antibody for its antigen. We conclude that IHC methodology optimization and validation are crucial steps for each antibody and each research question.


Subject(s)
Frozen Sections , Immunohistochemistry , Paraffin Embedding , Staining and Labeling , Antigens/metabolism , Biomarkers/analysis , Formaldehyde/pharmacology , Frozen Sections/methods , Humans , Immunohistochemistry/methods , Paraffin , Staining and Labeling/methods , Tissue Fixation/methods
7.
FASEB J ; 33(1): 557-571, 2019 01.
Article in English | MEDLINE | ID: mdl-30001166

ABSTRACT

Diffuse gliomas often carry point mutations in isocitrate dehydrogenase ( IDH1mut), resulting in metabolic stress. Although IDHmut gliomas are difficult to culture in vitro, they thrive in the brain via diffuse infiltration, suggesting brain-specific tumor-stroma interactions that can compensate for IDH-1 deficits. To elucidate the metabolic adjustments in clinical IDHmut gliomas that contribute to their malignancy, we applied a recently developed method of targeted quantitative RNA next-generation sequencing to 66 clinical gliomas and relevant orthotopic glioma xenografts, with and without the endogenous IDH-1R132H mutation. Datasets were analyzed in R using Manhattan plots to calculate distance between expression profiles, Ward's method to perform unsupervised agglomerative clustering, and the Mann Whitney U test and Fisher's exact tests for supervised group analyses. The significance of transcriptome data was investigated by protein analysis, in situ enzymatic activity mapping, and in vivo magnetic resonance spectroscopy of orthotopic IDH1mut- and IDHwt-glioma xenografts. Gene set enrichment analyses of clinical IDH1mut gliomas strongly suggest a role for catabolism of lactate and the neurotransmitter glutamate, whereas, in IDHwt gliomas, processing of glucose and glutamine are the predominant metabolic pathways. Further evidence of the differential metabolic activity in these cancers comes from in situ enzymatic mapping studies and preclinical in vivo magnetic resonance spectroscopy imaging. Our data support an evolutionary model in which IDHmut glioma cells exist in symbiosis with supportive neuronal cells and astrocytes as suppliers of glutamate and lactate, possibly explaining the diffuse nature of these cancers. The dependency on glutamate and lactate opens the way for novel approaches in the treatment of IDHmut gliomas.-Lenting, K., Khurshed, M., Peeters, T. H., van den Heuvel, C. N. A. M., van Lith, S. A. M., de Bitter, T., Hendriks, W., Span, P. N., Molenaar, R. J., Botman, D., Verrijp, K., Heerschap, A., ter Laan, M., Kusters, B., van Ewijk, A., Huynen, M. A., van Noorden, C. J. F., Leenders, W. P. J. Isocitrate dehydrogenase 1-mutated human gliomas depend on lactate and glutamate to alleviate metabolic stress.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Glutamic Acid/metabolism , Isocitrate Dehydrogenase/genetics , Lactic Acid/metabolism , Mutation , Stress, Physiological , 4-Aminobutyrate Transaminase/genetics , 4-Aminobutyrate Transaminase/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Glutaminase/genetics , Glutaminase/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Succinate-Semialdehyde Dehydrogenase/genetics , Succinate-Semialdehyde Dehydrogenase/metabolism , Transcriptome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Mol Histol ; 49(5): 481-497, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30046941

ABSTRACT

Glioblastoma (GBM) is the most lethal brain tumor also due to malignant and therapy-resistant GBM stem cells (GSCs) that are localized in protecting hypoxic GSC niches. Some members of the cysteine cathepsin family of proteases have been found to be upregulated in GBM. Cathepsin K gene expression is highly elevated in GBM tissue versus normal brain and it has been suggested to regulate GSC migration out of the niches. Here, we investigated the cellular distribution of cathepsins B, X and K in GBM tissue and whether these cathepsins are co-localized in GSC niches. Therefore, we determined expression of these cathepsins in serial paraffin sections of 14 human GBM samples and serial cryostat sections of two samples using immunohistochemistry and metabolic mapping of cathepsin activity using selective fluorogenic substrates. We detected cathepsins B, X and K in peri-arteriolar GSC niches in 9 out of 16 GBM samples, which were defined by co-expression of the GSC marker CD133, the niche marker stromal-derived factor-1α (SDF-1α) and smooth muscle actin as a marker for arterioles. The expression of cathepsin B and X was detected in stromal cells and cancer cells throughout the GBM sections, whereas cathepsin K expression was more restricted to arteriole-rich regions in the GBM sections. Metabolic mapping showed that cathepsin B, but not cathepsin K is active in GSC niches. On the basis of these findings, it is concluded that cathepsins B, X and K have distinct functions in GBM and that cathepsin K is the most likely GSC niche-related cathepsin of the three cathepsins investigated.


Subject(s)
Cathepsins/metabolism , Glioblastoma/pathology , Stem Cell Niche , Adult , Aged , Aged, 80 and over , Arterioles , Cathepsin B/analysis , Cathepsin B/metabolism , Cathepsin K , Cathepsin Z/analysis , Cathepsin Z/metabolism , Cathepsins/analysis , Glioblastoma/enzymology , Glioblastoma/metabolism , Humans , Immunohistochemistry , Middle Aged , Proteolysis
9.
J Vis Exp ; (135)2018 05 26.
Article in English | MEDLINE | ID: mdl-29889193

ABSTRACT

Altered cellular metabolism is a hallmark of many diseases, including cancer, cardiovascular diseases and infection. The metabolic motor units of cells are enzymes and their activity is heavily regulated at many levels, including the transcriptional, mRNA stability, translational, post-translational and functional level. This complex regulation means that conventional quantitative or imaging assays, such as quantitative mRNA experiments, Western Blots and immunohistochemistry, yield incomplete information regarding the ultimate activity of enzymes, their function and/or their subcellular localization. Quantitative enzyme cytochemistry and histochemistry (i.e., metabolic mapping) show in-depth information on in situ enzymatic activity and its kinetics, function and subcellular localization in an almost true-to-nature situation. We describe a protocol to detect the activity of dehydrogenases, which are enzymes that perform redox reactions to reduce cofactors such as NAD(P)+ and FAD. Cells and tissue sections are incubated in a medium that is specific for the enzymatic activity of one dehydrogenase. Subsequently, the dehydrogenase that is the subject of investigation performs its enzymatic activity in its subcellular site. In a chemical reaction with the reaction medium, this ultimately generates blue-colored formazan at the site of the dehydrogenase's activity. The formazan's absorbance is therefore a direct measure of the dehydrogenase's activity and can be quantified using monochromatic light microscopy and image analysis. The quantitative aspect of this protocol enables researchers to draw statistical conclusions from these assays. Besides observational studies, this technique can be used for inhibition studies of specific enzymes. In this context, studies benefit from the true-to-nature advantages of metabolic mapping, giving in situ results that may be physiologically more relevant than in vitro enzyme inhibition studies. In all, metabolic mapping is an indispensable technique to study metabolism at the cellular or tissue level. The technique is easy to adopt, provides in-depth, comprehensive and integrated metabolic information and enables rapid quantitative analysis.


Subject(s)
Histocytochemistry/methods , Immunohistochemistry/methods , Oxidoreductases/chemistry , Humans
10.
FASEB J ; : fj201800547R, 2018 Jun 07.
Article in English | MEDLINE | ID: mdl-29879375

ABSTRACT

Isocitrate dehydrogenase ( IDH1)-1 is mutated in various types of human cancer, and the presence of this mutation is associated with improved responses to irradiation and chemotherapy in solid tumor cells. Mutated IDH1 (IDH1MUT) enzymes consume NADPH to produce d-2-hydroxyglutarate (d-2HG) resulting in the decreased reducing power needed for detoxification of reactive oxygen species (ROS), for example. The objective of the current study was to investigate the mechanism behind the chemosensitivity of the widely used anticancer agent cisplatin in IDH1MUT cancer cells. Oxidative stress, DNA damage, and mitochondrial dysfunction caused by cisplatin treatment were monitored in IDH1MUT HCT116 colorectal cancer cells and U251 glioma cells. We found that exposure to cisplatin induced higher levels of ROS, DNA double-strand breaks (DSBs), and cell death in IDH1MUT cancer cells, as compared with IDH1 wild-type ( IDH1WT) cells. Mechanistic investigations revealed that cisplatin treatment dose dependently reduced oxidative respiration in IDH1MUT cells, which was accompanied by disturbed mitochondrial proteostasis, indicative of impaired mitochondrial activity. These effects were abolished by the IDH1MUT inhibitor AGI-5198 and were restored by treatment with d-2HG. Thus, our study shows that altered oxidative stress responses and a vulnerable oxidative metabolism underlie the sensitivity of IDH1MUT cancer cells to cisplatin.-Khurshed, M., Aarnoudse, N., Hulsbos, R., Hira, V. V. V., van Laarhoven, H. W. M., Wilmink, J. W., Molenaar, R. J., van Noorden, C. J. F. IDH1-mutant cancer cells are sensitive to cisplatin and an IDH1-mutant inhibitor counteracts this sensitivity.

11.
Arthropod Struct Dev ; 47(2): 199-213, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29421154

ABSTRACT

Calcium bodies are internal epithelial sacs found in terrestrial isopods of the family Trichoniscidae that contain a mineralized extracellular matrix that is deposited and resorbed in relation to the molt cycle. Calcium bodies in several trichoniscids are filled with bacteria, the function of which is currently unknown. The woodlouse Hyloniscus riparius differs from other trichoniscids in that it possesses two different pairs of calcium bodies, the posterior pair being filled with bacteria and the anterior pair being devoid of bacteria. We explored the development of these organs and bacterial colonization of their lumen during the postmarsupial development with the use of optical clearing and whole-body confocal imaging of larval and juvenile stages. Our results show that calcium bodies are formed as invaginations of the epidermis in the region of intersegmental membranes during the postmarsupial development. The anterior pair of calcium bodies is generated during the first postmarsupial manca stage, whereas the posterior calcium bodies first appear in juveniles and are immediately colonized by bacteria, likely through a connection between the calcium body lumen and the body surface. Mineral is deposited in calcium bodies as soon as they are present.


Subject(s)
Calcium/metabolism , Extracellular Matrix/physiology , Isopoda/growth & development , Molting , Animals , Epidermis/growth & development , Epidermis/ultrastructure , Extracellular Matrix/ultrastructure , Isopoda/ultrastructure , Larva/growth & development , Larva/ultrastructure , Microscopy, Electron, Scanning
12.
Clin Cancer Res ; 24(7): 1705-1715, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29339439

ABSTRACT

Purpose: Somatic mutations in IDH1/2 occur in approximately 20% of patients with myeloid neoplasms, including acute myeloid leukemia (AML). IDH1/2MUT enzymes produce D-2-hydroxyglutarate (D2HG), which associates with increased DNA damage and improved responses to chemo/radiotherapy and PARP inhibitors in solid tumor cells. Whether this also holds true for IDH1/2MUT AML is not known.Experimental Design: Well-characterized primary IDH1MUT, IDH2MUT, and IDH1/2WT AML cells were analyzed for DNA damage and responses to daunorubicin, ionizing radiation, and PARP inhibitors.Results:IDH1/2MUT caused increased DNA damage and sensitization to daunorubicin, irradiation, and the PARP inhibitors olaparib and talazoparib in AML cells. IDH1/2MUT inhibitors protected against these treatments. Combined treatment with a PARP inhibitor and daunorubicin had an additive effect on the killing of IDH1/2MUT AML cells. We provide evidence that the therapy sensitivity of IDH1/2MUT cells was caused by D2HG-mediated downregulation of expression of the DNA damage response gene ATM and not by altered redox responses due to metabolic alterations in IDH1/2MUT cells.Conclusions:IDH1/2MUT AML cells are sensitive to PARP inhibitors as monotherapy but especially when combined with a DNA-damaging agent, such as daunorubicin, whereas concomitant administration of IDH1/2MUT inhibitors during cytotoxic therapy decrease the efficacy of both agents in IDH1/2MUT AML. These results advocate in favor of clinical trials of PARP inhibitors either or not in combination with daunorubicin in IDH1/2MUT AML. Clin Cancer Res; 24(7); 1705-15. ©2018 AACR.


Subject(s)
Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation/drug effects , Mutation/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/genetics , Daunorubicin/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , HCT116 Cells , Humans , Oxidation-Reduction/drug effects , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerases/metabolism
13.
J Histochem Cytochem ; 66(3): 155-173, 2018 03.
Article in English | MEDLINE | ID: mdl-29297738

ABSTRACT

In glioblastoma, a fraction of malignant cells consists of therapy-resistant glioblastoma stem cells (GSCs) residing in protective niches that recapitulate hematopoietic stem cell (HSC) niches in bone marrow. We have previously shown that HSC niche proteins stromal cell-derived factor-1α (SDF-1α), C-X-C chemokine receptor type 4 (CXCR4), osteopontin (OPN), and cathepsin K (CatK) are expressed in hypoxic GSC niches around arterioles in five human glioblastoma samples. In HSC niches, HSCs are retained by binding of SDF-1α and OPN to their receptors CXCR4 and CD44, respectively. Protease CatK cleaves SDF-1α to release HSCs out of niches. The aim of the present study was to reproduce the immunohistochemical localization of these GSC markers in 16 human glioblastoma samples with the addition of three novel markers. Furthermore, we assessed the type of blood vessels associated with GSC niches. In total, we found seven GSC niches containing CD133-positive and nestin-positive GSCs as a single-cell layer exclusively around the tunica adventitia of 2% of the CD31-positive and SMA-positive arterioles and not around capillaries and venules. Niches expressed SDF-1α, CXCR4, CatK, OPN, CD44, hypoxia-inducible factor-1α, and vascular endothelial growth factor. In conclusion, we show that GSC niches are present around arterioles and express bone marrow HSC niche proteins.


Subject(s)
Arterioles/pathology , Brain Neoplasms/pathology , Glioblastoma/pathology , Hematopoietic Stem Cells/pathology , Neoplastic Stem Cells/pathology , Stem Cell Niche , Adult , Aged , Brain Neoplasms/blood supply , Cathepsin K/analysis , Chemokine CXCL12/analysis , Glioblastoma/blood supply , Humans , Hyaluronan Receptors/analysis , Immunohistochemistry/methods , Middle Aged , Osteopontin/analysis , Receptors, CXCR4/analysis , Staining and Labeling/methods
14.
Sci Rep ; 8(1): 1647, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29374186

ABSTRACT

In the present study, 3D histochemistry and imaging methodology is described for human gingiva to analyze its vascular network. Fifteen human gingiva samples without signs of inflammation were cleared using a mixture of 2-parts benzyl benzoate and 1-part benzyl alcohol (BABB), after being immunofluorescently stained for CD31, marker of endothelial cells to visualize blood vessels in combination with fluorescent DNA dyes. Samples were imaged in 3D with the use of confocal microscopy and light-sheet microscopy and image processing. BABB clearing caused limited tissue shrinkage 13 ± 7% as surface area and 24 ± 1% as volume. Fluorescence remained intact in BABB-cleared gingiva samples and light-sheet microscopy was an excellent tool to image gingivae whereas confocal microscopy was not. Histochemistry on cryostat sections of gingiva samples after 3D imaging validated structures visualized in 3D. Three-dimensional images showed the vascular network in the stroma of gingiva with one capillary loop in each stromal papilla invading into the epithelium. The capillary loops were tortuous with structural irregularities that were not apparent in 2D images. It is concluded that 3D histochemistry and imaging methodology described here is a promising novel approach to study structural aspects of human gingiva in health and disease.


Subject(s)
Blood Vessels/anatomy & histology , Gingiva/anatomy & histology , Histocytochemistry/methods , Imaging, Three-Dimensional/methods , Optical Imaging/methods , Endothelial Cells/chemistry , Humans , Microscopy , Microscopy, Confocal , Microscopy, Fluorescence , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Staining and Labeling/methods
15.
BMJ Open ; 7(6): e014961, 2017 06 10.
Article in English | MEDLINE | ID: mdl-28601826

ABSTRACT

INTRODUCTION: High-grade chondrosarcoma, high-grade glioma and intrahepatic cholangiocarcinoma are aggressive types of cancer with a dismal outcome. This is due to the lack of effective treatment options, emphasising the need for novel therapies. Mutations in the genes IDH1 and IDH2 (isocitrate dehydrogenase 1 and 2) occur in 60% of chondrosarcoma, 80% of WHO grade II-IV glioma and 20% of intrahepatic cholangiocarcinoma. IDH1/2-mutated cancer cells produce the oncometabolite D-2-hydroxyglutarate (D-2HG) and are metabolically vulnerable to treatment with the oral antidiabetic metformin and the oral antimalarial drug chloroquine. METHODS AND ANALYSIS: We describe a dose-finding phase Ib/II clinical trial, in which patients with IDH1/2-mutated chondrosarcoma, glioma and intrahepatic cholangiocarcinoma are treated with a combination of metformin and chloroquine. Dose escalation is performed according to a 3+3 dose-escalation scheme. The primary objective is to determine the maximum tolerated dose to establish the recommended dose for a phase II clinical trial. Secondary objectives of the study include (1) determination of pharmacokinetics and toxic effects of the study therapy, for which metformin and chloroquine serum levels will be determined over time; (2) investigation of tumour responses to metformin plus chloroquine in IDH1/2-mutated cancers using CT/MRI scans; and (3) whether tumour responses can be measured by non-invasive D-2HG measurements (mass spectrometry and magnetic resonance spectroscopy) of tumour tissue, serum, urine, and/or bile or next-generation sequencing of circulating tumour DNA (liquid biopsies). This study may open a novel treatment avenue for IDH1/2-mutated high-grade chondrosarcoma, glioma and intrahepatic cholangiocarcinoma by repurposing the combination of two inexpensive drugs that are already approved for other indications. ETHICS AND DISSEMINATION: This study has been approved by the medical-ethical review committee of the Academic Medical Center, Amsterdam, The Netherlands. The report will be submitted to a peer-reviewed journal. TRIAL REGISTRATION NUMBER: This article was registered at ClinicalTrials.gov identifier (NCT02496741): Pre-results.


Subject(s)
Antineoplastic Agents/therapeutic use , Chloroquine/therapeutic use , Cholangiocarcinoma/drug therapy , Chondrosarcoma/drug therapy , Glioma/drug therapy , Metformin/therapeutic use , Adult , Antineoplastic Agents/pharmacokinetics , Chloroquine/pharmacokinetics , Cholangiocarcinoma/genetics , Chondrosarcoma/genetics , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Male , Metformin/pharmacokinetics , Research Design
16.
Oncotarget ; 8(30): 49165-49177, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28467784

ABSTRACT

Hotspot mutations in isocitrate dehydrogenase 1 (IDH1) initiate low-grade glioma and secondary glioblastoma and induce a neomorphic activity that converts α-ketoglutarate (α-KG) to the oncometabolite D-2-hydroxyglutarate (D-2-HG). It causes metabolic rewiring that is not fully understood. We investigated the effects of IDH1 mutations (IDH1MUT) on expression of genes that encode for metabolic enzymes by data mining The Cancer Genome Atlas. We analyzed 112 IDH1 wild-type (IDH1WT) versus 399 IDH1MUT low-grade glioma and 157 IDH1WT versus 9 IDH1MUT glioblastoma samples. In both glioma types, IDH1WT was associated with high expression levels of genes encoding enzymes that are involved in glycolysis and acetate anaplerosis, whereas IDH1MUT glioma overexpress genes encoding enzymes that are involved in the oxidative tricarboxylic acid (TCA) cycle. In vitro, we observed that IDH1MUT cancer cells have a higher basal respiration compared to IDH1WT cancer cells and inhibition of the IDH1MUT shifts the metabolism by decreasing oxygen consumption and increasing glycolysis. Our findings indicate that IDH1WT glioma have a typical Warburg phenotype whereas in IDH1MUT glioma the TCA cycle, rather than glycolytic lactate production, is the predominant metabolic pathway. Our data further suggest that the TCA in IDH1MUT glioma is driven by lactate and glutamate anaplerosis to facilitate production of α-KG, and ultimately D-2-HG. This metabolic rewiring may be a basis for novel therapies for IDH1MUT and IDH1WT glioma.


Subject(s)
Glioma/genetics , Glioma/metabolism , Glutamic Acid/metabolism , Isocitrate Dehydrogenase/genetics , Lactic Acid/metabolism , Mutation , Citric Acid Cycle/genetics , Computational Biology/methods , Databases, Genetic , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioma/pathology , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glycolysis/genetics , Humans , Metabolic Networks and Pathways , Oxygen/metabolism , Phenotype , Transcriptome
17.
Cancer Res ; 75(22): 4790-802, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26363012

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is mutated in various types of human cancer to IDH1(R132H), a structural alteration that leads to catalysis of α-ketoglutarate to the oncometabolite D-2-hydroxyglutarate. In this study, we present evidence that small-molecule inhibitors of IDH1(R132H) that are being developed for cancer therapy may pose risks with coadministration of radiotherapy. Cancer cells heterozygous for the IDH1(R132H) mutation exhibited less IDH-mediated production of NADPH, such that after exposure to ionizing radiation (IR), there were higher levels of reactive oxygen species, DNA double-strand breaks, and cell death compared with IDH1 wild-type cells. These effects were reversed by the IDH1(R132H) inhibitor AGI-5198. Exposure of IDH1 wild-type cells to D-2-hydroxyglutarate was sufficient to reduce IDH-mediated NADPH production and increase IR sensitivity. Mechanistic investigations revealed that the radiosensitivity of heterozygous cells was independent of the well-described DNA hypermethylation phenotype in IDH1-mutated cancers. Thus, our results argue that altered oxidative stress responses are a plausible mechanism to understand the radiosensitivity of IDH1-mutated cancer cells. Further, they offer an explanation for the relatively longer survival of patients with IDH1-mutated tumors, and they imply that administration of IDH1(R132H) inhibitors in these patients may limit irradiation efficacy in this setting.


Subject(s)
Antineoplastic Agents/pharmacology , Benzeneacetamides/pharmacology , Glioblastoma/genetics , Imidazoles/pharmacology , Isocitrate Dehydrogenase/genetics , Radiation Tolerance/drug effects , Blotting, Western , Cell Line, Tumor , Chemoradiotherapy/adverse effects , DNA Methylation/drug effects , DNA Methylation/radiation effects , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Gene Knock-In Techniques , Glioblastoma/pathology , Humans , In Vitro Techniques , Mutation , NADP/biosynthesis , Oxidative Stress/drug effects , Oxidative Stress/radiation effects
18.
Asian Pac J Cancer Prev ; 9(3): 533-6, 2008.
Article in English | MEDLINE | ID: mdl-18990034

ABSTRACT

INTRODUCTION: The objective of the study was to determine the trends of cancer cervix in Karachi South during an eight (1995-2002) year period. METHODOLOGY: Cancer cervix cases recorded at Karachi Cancer Registry during 1st January 1995 to 31st December 2002 were analyzed. Trends were studied by analyzing the age standardized incidence rates (ASR)s in 2 time periods, 1995-97 and 1998-2002. RESULTS: Cancer cervix ranked sixth in the 1995-97 period the age standardized incidence rate (ASR) world and crude incidence rate (CIR) per 100,000 were 6.81 and 3.22. It reached the fifth ranking in the 1998-2002 period with an ASR and CIR of 7.5 and 4.0 per 100,000. Thus between 1995 and 2002, the incidence of cervical cancer registered an approximate 10% increase. The mean age of the cancer cases was 53.3 years (SD 11.6; 95% CI 50.58, 55.96; range 32-85 years) and 50.7 years (SD 11.7; 95% CI 48.8, 52.5; range 51 years) in period 1 and 2 respectively. The morphological components of squamous cell carcinoma and adenocarcinoma remained stable during this period, though a marginally higher component and increasing incidence of adenocarcinoma was observed throughout. A negligible down staging was observed in the 1998-2002 period. Localized malignancy was observed in 30.8% in period 2 as compared to 25.7% in period 1 and the component of carcinoma in situ increased from 0% percent in period 1 to 1.3% in the second period. Despite this two thirds of the cases still presented with a regional or distant spread of disease. CONCLUSION: Pakistan at present falls into a low risk cancer cervix region. The cause of concern is the steadily increasing incidence especially in the younger birth cohorts, the advanced disease at presentation; insignificant in-situ cancers and no preventive intervention or awareness practices in place.


Subject(s)
Neoplasm Invasiveness/pathology , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/pathology , Adult , Age Distribution , Aged , Developing Countries , Female , Humans , Incidence , Neoplasm Staging , Pakistan/epidemiology , Registries , Retrospective Studies , Risk Assessment , Survival Analysis , Uterine Cervical Neoplasms/ethnology
19.
Asian Pac J Cancer Prev ; 9(3): 453-8, 2008.
Article in English | MEDLINE | ID: mdl-19004134

ABSTRACT

OBJECTIVE: The primary objective of this study was to analyze the anatomic distribution, clinical features and outcome of Diffuse large B-cell lymphoma (DLBCL) patients according to the primary site (extranodal vs. nodal) with applicability of International Prognostic Index (IPI). METHODOLOGY: A retrospective review (1988 to 2004) of 557 cases of DLBC. RESULTS: The median age was 48.7 +/- 15.3 years; M:F ratio was 2:1. The distribution according to the primary site was: lymph node (N-NHL), 322 cases (58%) of which 145(44%) were stage IV, 76(23%) stage III, 60 (18%) stage II and 47 (15%) stage I. The extra nodal sites (EN-NHL) 235 (42%) cases included gastro-intestinal tract (44%), upper aerodigestive tract (19%), bones (8%), spine (5%), and unusual sites less than 3% each as breast, CNS, testis, lungs and skin. The median survival rate was 4.8 years and 6.3 years in N-NHL and EN-NHL respectively. In the latter this varied greatly depending on the primary site and stage of disease at presentation. In the univariate analysis factors associated with good prognosis were: age less than 60 years, early stage (I-II), extranodal involvement primarily gastric or bone, 0-1 extranodal site, 0-1 performance status, lack of B symptoms and normal LDH level. In the multivariate analysis age, performance status, stage of disease and level of LDH were the main variables predicting overall survival; no nodal or extranodal site maintained their prognostic value. CONCLUSION: Patients with EN-NHL present more frequently with early stage disease then those with N-NHL; overall survival in both groups largely depended on IPI and not on the site of origin of the malignancy.


Subject(s)
Lymph Nodes/pathology , Lymphoma, Extranodal NK-T-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Neoplasm Invasiveness/pathology , Adult , Analysis of Variance , Biopsy, Needle , Cohort Studies , Confidence Intervals , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphoma, Extranodal NK-T-Cell/mortality , Lymphoma, Extranodal NK-T-Cell/therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Non-Hodgkin/mortality , Lymphoma, Non-Hodgkin/pathology , Lymphoma, Non-Hodgkin/therapy , Male , Middle Aged , Odds Ratio , Pakistan , Probability , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Assessment , Survival Analysis
20.
Asian Pac J Cancer Prev ; 9(2): 303-8, 2008.
Article in English | MEDLINE | ID: mdl-18712980

ABSTRACT

INTRODUCTION: Febrile neutropenia (FN) is a major complication of chemotherapy, costly in terms of morbidity, mortality and associated financial expenditure. The present study was conducted with the goal of highlighting FN as a serious problem in Pakistan, with the longer term objective of improved cancer survival, reduction in length of stay (LOS) in hospital, morbidity, mortality and costs in our existing developing country scenario. METHODS: A cross-sectional descriptive study was conducted on patients, > or =18 years, admitted with FN as a consequence of chemotherapy at a referral hospital in Karachi from 1st September 2006 to 30th April 2007. RESULTS: A total of 80 patients [43 (53.8%) males and 37 (46.2%) females] were selected. The mean age was 47.4 (SD +/-16.6; range 18-79) years. Sixty eight patients (86%) were < or = 65 years, 50% were < or = 50 years. Overall, inhospital mortality was 11%; 4% for patients on granulocyte colony stimulating factor (G-CSF) prophylaxis as against 20% for those without. The cause of death was either pneumonia or septic shock. Mean LOS was 7.53 (SD +/-3.8; range 2-17) days. Hematological malignancies, older age, severity of dehydration, pneumonia and culture positivity were significantly associated with LOS and death. Those above 50 years of age were 1.5 times as likely to be hospitalized longer and > three times as likely to die. Bacteremia conferred a 5-fold and pneumonia an 8-fold increase in the risk of death. CONCLUSION: The results of this study indicate that age, vital instability, dehydration, high creatinine, culture positivity and hematological malignancies are high risk factors in chemotherapy induced FN. Identification of FN risk factors with poor outcomes may help in devising protocols for modified dosage or including GCFs initially. This may help reduce the cost of cancer care as well as mortality and morbidity. Prospective studies of FN in multiple centers in Pakistan may be beneficial in evaluating these risk factors further.


Subject(s)
Hospital Mortality/trends , Hospitalization/statistics & numerical data , Length of Stay , Neoplasms/drug therapy , Neutropenia/chemically induced , Neutropenia/mortality , Adolescent , Adult , Aged , Cross-Sectional Studies , Female , Fever/chemically induced , Fever/mortality , Health Care Costs , Humans , Male , Middle Aged , Risk Factors , Survival Rate , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...