Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Thyroid ; 29(7): 979-992, 2019 07.
Article in English | MEDLINE | ID: mdl-30938231

ABSTRACT

Background: Anaplastic thyroid carcinoma (ATC) is one of the most aggressive human cancers, with a median survival of only three to six months. Standard treatment options and even targeted therapies have so far failed to improve long-term overall survival. Thus, novel treatment modalities for ATC, such as immunotherapy, are urgently needed. CD47 is a "don't eat me" signal, which prevents cancer cells from phagocytosis by binding to signal regulatory protein alpha on macrophages. So far, the role of macrophages and the CD47-signal regulatory protein alpha signaling axis in ATC is not well understood. Methods: This study analyzed 19 primary human ATCs for macrophage markers, CD47 expression, and immune checkpoints by immunohistochemistry. ATC cell lines and a fresh ATC sample were assessed by flow cytometry for CD47 expression and macrophage infiltration, respectively. CD47 was blocked in phagocytosis assays of co-cultured macrophages and ATC cell lines. Anti-CD47 antibody treatment was administered to ATC cell line xenotransplanted immunocompromised mice, as well as to tamoxifen-induced ATC double-transgenic mice. Results: Human ATC samples were heavily infiltrated by CD68- and CD163-expressing tumor-associated macrophages (TAMs), and expressed CD47 and calreticulin, the dominant pro-phagocytic molecule. In addition, ATC tissues expressed the immune checkpoint molecules programmed cell death 1 and programmed death ligand 1. Blocking CD47 promoted the phagocytosis of ATC cell lines by macrophages in vitro. Anti-CD47 antibody treatment of ATC xenotransplanted mice increased the frequency of TAMs, enhanced the expression of macrophage activation markers, augmented tumor cell phagocytosis, and suppressed tumor growth. In double-transgenic ATC mice, CD47 was expressed on tumor cells, and blocking CD47 increased TAM frequencies. Conclusions: Targeting CD47 or CD47 in combination with programmed cell death 1 may potentially improve the outcomes of ATC patients and may represent a valuable addition to the current standard of care.


Subject(s)
Antigens, Differentiation/immunology , CD47 Antigen/immunology , Macrophages/immunology , Phagocytosis/immunology , Receptors, Immunologic/immunology , Thyroid Carcinoma, Anaplastic/immunology , Thyroid Neoplasms/immunology , Tumor Escape/immunology , Aged , Aged, 80 and over , Animals , Antigens, Differentiation/metabolism , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/metabolism , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Immunotherapy , In Vitro Techniques , Macrophages/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Molecular Targeted Therapy , Neoplasm Transplantation , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/metabolism , Xenograft Model Antitumor Assays
2.
J Nucl Med ; 60(7): 917-923, 2019 07.
Article in English | MEDLINE | ID: mdl-30464041

ABSTRACT

Anaplastic thyroid carcinoma (ATC) is refractory to radioiodine therapy in part because of impaired iodine metabolism. We targeted the mitogen-activated protein kinase and phosphatidylinositol 3-kinase (PI3'K) pathways with the intent to induce radioiodine uptake for radioiodine treatment of ATC. Methods: Human ATC cells were used to evaluate the ability of pharmacologic inhibition of the mitogen-activated protein kinase and PI3'K pathways to induce radioiodine uptake. Thyrocyte-specific double-mutant BRAFV600E PIK3CAH1047R mice were treated with a MEK inhibitor followed by radioiodine treatment, and tumor burden was monitored by ultrasound imaging. Results: ATC cell lines showed an increase in sodium-iodine symporter transcription when treated with a MEK or BRAFV600E inhibitor alone and in combination with PI3'K inhibitor. This translated into a dose-dependent elevation of iodine uptake after treatment with a MEK inhibitor alone and in combination with a PI3'K inhibitor. In vivo, MEK inhibition but not BRAF or PI3'K inhibition upregulated sodium-iodine symporter transcription. This translated into a stable reduction of tumor burden when mice were treated with a MEK inhibitor before radioiodine administration. Conclusion: This study confirms the ability of MEK inhibition to induce iodine uptake in in vitro and in vivo models of ATC. The approach of using a MEK inhibitor before radioiodine treatment could readily be translated into clinical practice and provide a much-needed therapeutic option for patients with ATC.


Subject(s)
Iodine Radioisotopes/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/metabolism , Animals , Biological Transport/drug effects , Cell Line, Tumor , Disease Models, Animal , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Iodine Radioisotopes/therapeutic use , Mice , RNA, Messenger/genetics , Symporters/genetics , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Carcinoma, Anaplastic/radiotherapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/radiotherapy , Transcription, Genetic/drug effects
3.
ChemMedChem ; 14(2): 224-236, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30520265

ABSTRACT

By screening a focused library of kinase inhibitor analogues in a phenotypic co-culture assay for angiogenesis inhibition, we identified an aminotriazine that acts as a cytostatic nanomolar inhibitor. However, this aminotriazine was found to be completely inactive in a whole-kinome profiling assay. To decipher its mechanism of action, we used the online target prediction tool PPB2 (http://ppb2.gdb.tools), which suggested lysophosphatidic acid acyltransferaseâ€…ß (LPAAT-ß) as a possible target for this aminotriazine as well as several analogues identified by structure-activity relationship profiling. LPAAT-ß inhibition (IC50 ≈15 nm) was confirmed in a biochemical assay and by its effects on cell proliferation in comparison with a known LPAAT-ß inhibitor. These experiments illustrate the value of target-prediction tools to guide target identification for phenotypic screening hits and significantly expand the rather limited pharmacology of LPAAT-ß inhibitors.


Subject(s)
Acyltransferases/antagonists & inhibitors , Angiogenesis Inducing Agents/metabolism , Enzyme Inhibitors/chemistry , Small Molecule Libraries/chemistry , Triazines/chemistry , Acyltransferases/genetics , Acyltransferases/isolation & purification , Biological Assay/methods , Cell Culture Techniques , Cell Line , Cell Proliferation/drug effects , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Phenotype , Protein Binding , Small Molecule Libraries/metabolism , Software , Structure-Activity Relationship , Triazines/metabolism
4.
Oncotarget ; 8(61): 103207-103222, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262556

ABSTRACT

Thyroid carcinomas are the most prevalent endocrine cancers. The BRAFV600E mutation is found in 40% of the papillary type and 25% of the anaplastic type. BRAFV600E inhibitors have shown great success in melanoma but, they have been, to date, less successful in thyroid cancer. About 50% of anaplastic thyroid carcinomas present mutations/amplification of the phosphatidylinositol 3' kinase. Here we propose to investigate if the hyper activation of that pathway could influence the response to BRAFV600E specific inhibitors. To test this, we used two mouse models of thyroid cancer. Single mutant (BRAFV600E) mice responded to BRAFV600E-specific inhibition (PLX-4720), while double mutant mice (BRAFV600E; PIK3CAH1047R) showed resistance and even signs of aggravation. This resistance was abrogated by combination with a phosphoinositide 3-kinase inhibitor. At the molecular level, we showed that this resistance was concomitant to a paradoxical activation of the MAP-Kinase pathway, which could be overturned by phosphoinositide 3-kinase inhibition in vivo in our mouse model and in vitro in human double mutant cell lines. In conclusion, we reveal a phosphoinositide 3-kinase driven, paradoxical MAP-Kinase pathway activation as mechanism for resistance to BRAFV600E specific inhibitors in a clinically relevant mouse model of thyroid cancer.

5.
Oncotarget ; 8(15): 24604-24620, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445948

ABSTRACT

Anaplastic thyroid cancers and radioiodine resistant thyroid cancer are posing a major treat since surgery combined with Iodine131 therapy is ineffective on them. Small-molecule inhibitors are presenting a new hope for patients, but often lead to drug resistance in many cancers. Based on the major mutations found in thyroid cancer, we propose the combination of a MEK inhibitor and a Pi3'-kinase inhibitor in pre-clinical models. We used human thyroid cancer cell lines and genetically engineered double mutant BRAFV600E PIK3CAH1047R mice to evaluate the effect of both inhibitors separately or in combination in terms of proliferation and signaling in vitro; tumor burden, histology, cell death induction and tumor markers expression in vivo. The combination of MEK and Pi'3-kinase inhibition shows a synergistic effect in term of proliferation and apoptosis induction through Survivin down-regulation in vitro. We show for the first time the effects of the combination of a MEK inhibitor and Pi3'-kinase inhibitor in a genetically engineered mouse model of aggressively lethal thyroid cancer. In fine, the two drugs cooperate to promote tumor shrinkage by inducing a proliferation arrest and an elevation of apoptosis in vivo. Moreover, a phenotypic reversion is also observed with a partial restoration of normal thyroid marker transcription, and thyroid cancer marker expression reduction.In conclusion, combination therapy of MEK and Pi3'-kinase inhibition synergizes to target double mutant thyroid cancer in vitro and in vivo. This multidrug approach could readily be translated into clinical practice and bring new perspectives for the treatment of incurable thyroid carcinoma.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Drug Synergism , Female , Humans , Mice , Thyroid Neoplasms/genetics
6.
PLoS Pathog ; 11(3): e1004760, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25786000

ABSTRACT

The coordinated exit of intracellular pathogens from host cells is a process critical to the success and spread of an infection. While phospholipases have been shown to play important roles in bacteria host cell egress and virulence, their role in the release of intracellular eukaryotic parasites is largely unknown. We examined a malaria parasite protein with phospholipase activity and found it to be involved in hepatocyte egress. In hepatocytes, Plasmodium parasites are surrounded by a parasitophorous vacuole membrane (PVM), which must be disrupted before parasites are released into the blood. However, on a molecular basis, little is known about how the PVM is ruptured. We show that Plasmodium berghei phospholipase, PbPL, localizes to the PVM in infected hepatocytes. We provide evidence that parasites lacking PbPL undergo completely normal liver stage development until merozoites are produced but have a defect in egress from host hepatocytes. To investigate this further, we established a live-cell imaging-based assay, which enabled us to study the temporal dynamics of PVM rupture on a quantitative basis. Using this assay we could show that PbPL-deficient parasites exhibit impaired PVM rupture, resulting in delayed parasite egress. A wild-type phenotype could be re-established by gene complementation, demonstrating the specificity of the PbPL deletion phenotype. In conclusion, we have identified for the first time a Plasmodium phospholipase that is important for PVM rupture and in turn for parasite exit from the infected hepatocyte and therefore established a key role of a parasite phospholipase in egress.


Subject(s)
Hepatocytes/microbiology , Malaria/enzymology , Phospholipases/metabolism , Protozoan Proteins/metabolism , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Hepatocytes/enzymology , Mice , Plasmodium berghei/enzymology , Plasmodium berghei/pathogenicity , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Vacuoles/enzymology , Vacuoles/microbiology
7.
Autophagy ; 9(4): 568-80, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23388496

ABSTRACT

Analyzing molecular determinants of Plasmodium parasite cell death is a promising approach for exploring new avenues in the fight against malaria. Three major forms of cell death (apoptosis, necrosis and autophagic cell death) have been described in multicellular organisms but which cell death processes exist in protozoa is still a matter of debate. Here we suggest that all three types of cell death occur in Plasmodium liver-stage parasites. Whereas typical molecular markers for apoptosis and necrosis have not been found in the genome of Plasmodium parasites, we identified genes coding for putative autophagy-marker proteins and thus concentrated on autophagic cell death. We characterized the Plasmodium berghei homolog of the prominent autophagy marker protein Atg8/LC3 and found that it localized to the apicoplast. A relocalization of PbAtg8 to autophagosome-like vesicles or vacuoles that appear in dying parasites was not, however, observed. This strongly suggests that the function of this protein in liver-stage parasites is restricted to apicoplast biology.


Subject(s)
Autophagy , Life Cycle Stages , Liver/parasitology , Parasites/cytology , Parasites/growth & development , Plasmodium berghei/cytology , Plasmodium berghei/growth & development , Amino Acid Sequence , Animals , Conserved Sequence , Databases, Protein , Evolution, Molecular , Gene Knockout Techniques , Genetic Complementation Test , Green Fluorescent Proteins/metabolism , Hep G2 Cells , Humans , Lipid Metabolism , Mice , Molecular Sequence Data , Parasites/ultrastructure , Phagosomes/metabolism , Phagosomes/ultrastructure , Plasmodium berghei/ultrastructure , Protein Transport , Protozoan Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Schizonts/cytology , Schizonts/metabolism , Schizonts/ultrastructure , Sequence Homology, Amino Acid , Vacuoles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...