Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(6): 114260, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38838225

ABSTRACT

Immunotherapy remains underexploited in acute myeloid leukemia (AML) compared to other hematological malignancies. Currently, gemtuzumab ozogamicin is the only therapeutic antibody approved for this disease. Here, to identify potential targets for immunotherapeutic intervention, we analyze the surface proteome of 100 genetically diverse primary human AML specimens for the identification of cell surface proteins and conduct single-cell transcriptome analyses on a subset of these specimens to assess antigen expression at the sub-population level. Through this comprehensive effort, we successfully identify numerous antigens and markers preferentially expressed by primitive AML cells. Many identified antigens are targeted by therapeutic antibodies currently under clinical evaluation for various cancer types, highlighting the potential therapeutic value of the approach. Importantly, this initiative uncovers AML heterogeneity at the surfaceome level, identifies several antigens and potential primitive cell markers characterizing AML subgroups, and positions immunotherapy as a promising approach to target AML subgroup specificities.


Subject(s)
Immunotherapy , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/pathology , Immunotherapy/methods , Membrane Proteins/metabolism
2.
EMBO Rep ; 25(3): 1469-1489, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38366255

ABSTRACT

Tumor acidosis is associated with increased invasiveness and drug resistance. Here, we take an unbiased approach to identify vulnerabilities of acid-exposed cancer cells by combining pH-dependent flow cytometry cell sorting from 3D colorectal tumor spheroids and transcriptomic profiling. Besides metabolic rewiring, we identify an increase in tetraploid cell frequency and DNA damage response as consistent hallmarks of acid-exposed cancer cells, supported by the activation of ATM and ATR signaling pathways. We find that regardless of the cell replication error status, both ATM and ATR inhibitors exert preferential growth inhibitory effects on acid-exposed cancer cells. The efficacy of a combination of these drugs with 5-FU is further documented in 3D spheroids as well as in patient-derived colorectal tumor organoids. These data position tumor acidosis as a revelator of the therapeutic potential of DNA repair blockers and as an attractive clinical biomarker to predict the response to a combination with chemotherapy.


Subject(s)
Colorectal Neoplasms , Tetraploidy , Humans , Ataxia Telangiectasia Mutated Proteins/genetics , Signal Transduction , DNA Damage , DNA Repair , Protein Kinase Inhibitors/pharmacology
3.
Blood Adv ; 8(1): 112-129, 2024 01 09.
Article in English | MEDLINE | ID: mdl-37729615

ABSTRACT

ABSTRACT: Acute megakaryoblastic leukemia (AMKL) is a rare, developmentally restricted, and highly lethal cancer of early childhood. The paucity and hypocellularity (due to myelofibrosis) of primary patient samples hamper the discovery of cell- and genotype-specific treatments. AMKL is driven by mutually exclusive chimeric fusion oncogenes in two-thirds of the cases, with CBFA2T3::GLIS2 (CG2) and NUP98 fusions (NUP98r) representing the highest-fatality subgroups. We established CD34+ cord blood-derived CG2 models (n = 6) that sustain serial transplantation and recapitulate human leukemia regarding immunophenotype, leukemia-initiating cell frequencies, comutational landscape, and gene expression signature, with distinct upregulation of the prosurvival factor B-cell lymphoma 2 (BCL2). Cell membrane proteomic analyses highlighted CG2 surface markers preferentially expressed on leukemic cells compared with CD34+ cells (eg, NCAM1 and CD151). AMKL differentiation block in the mega-erythroid progenitor space was confirmed by single-cell profiling. Although CG2 cells were rather resistant to BCL2 genetic knockdown or selective pharmacological inhibition with venetoclax, they were vulnerable to strategies that target the megakaryocytic prosurvival factor BCL-XL (BCL2L1), including in vitro and in vivo treatment with BCL2/BCL-XL/BCL-W inhibitor navitoclax and DT2216, a selective BCL-XL proteolysis-targeting chimera degrader developed to limit thrombocytopenia in patients. NUP98r AMKL were also sensitive to BCL-XL inhibition but not the NUP98r monocytic leukemia, pointing to a lineage-specific dependency. Navitoclax or DT2216 treatment in combination with low-dose cytarabine further reduced leukemic burden in mice. This work extends the cellular and molecular diversity set of human AMKL models and uncovers BCL-XL as a therapeutic vulnerability in CG2 and NUP98r AMKL.


Subject(s)
Antineoplastic Agents , Leukemia, Megakaryoblastic, Acute , Humans , Child , Child, Preschool , Animals , Mice , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Proteomics , Transcription Factors , Proto-Oncogene Proteins c-bcl-2 , Repressor Proteins
5.
J Exp Clin Cancer Res ; 41(1): 110, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35346305

ABSTRACT

BACKGROUND: CD44 is a multifunctional membrane glycoprotein. Through its heparan sulfate chain, CD44 presents growth factors to their receptors. We have shown that CD44 and Tropomyosin kinase A (TrkA) form a complex following nerve growth factor (NGF) induction. Our study aimed to understand how CD44 and TrkA interact and the consequences of inhibiting this interaction regarding the pro-tumoral effect of NGF in breast cancer. METHODS: After determining which CD44 isoforms (variants) are involved in forming the TrkA/CD44 complex using proximity ligation assays, we investigated the molecular determinants of this interaction. By molecular modeling, we isolated the amino acids involved and confirmed their involvement using mutations. A CD44v3 mimetic peptide was then synthesized to block the TrkA/CD44v3 interaction. The effects of this peptide on the growth, migration and invasion of xenografted triple-negative breast cancer cells were assessed. Finally, we investigated the correlations between the expression of the TrkA/CD44v3 complex in tumors and histo-pronostic parameters. RESULTS: We demonstrated that isoform v3 (CD44v3), but not v6, binds to TrkA in response to NGF stimulation. The final 10 amino acids of exon v3 and the TrkA H112 residue are necessary for the association of CD44v3 with TrkA. Functionally, the CD44v3 mimetic peptide impairs not only NGF-induced RhoA activation, clonogenicity, and migration/invasion of breast cancer cells in vitro but also tumor growth and metastasis in a xenograft mouse model. We also detected TrkA/CD44v3 only in cancerous cells, not in normal adjacent tissues. CONCLUSION: Collectively, our results suggest that blocking the CD44v3/TrkA interaction can be a new therapeutic option for triple-negative breast cancers.


Subject(s)
Breast Neoplasms , Hyaluronan Receptors , Nerve Growth Factor , Receptor, trkA , Animals , Breast Neoplasms/genetics , Female , Humans , Hyaluronan Receptors/metabolism , Mice , Nerve Growth Factor/pharmacology , Protein Isoforms , Receptor, trkA/metabolism
6.
Mol Cell Oncol ; 7(6): 1822123, 2020 Oct 07.
Article in English | MEDLINE | ID: mdl-33235918

ABSTRACT

KRAS-driven cancers acquire profound metabolic dependencies that are intimately linked to tumor growth. Our work revealed that colorectal cancers that harbor KRAS mutations are addicted to copper metabolism. This adaptation renders tumor cells critically dependent on the copper transporter ATP7A, which reveals copper metabolism as a promising therapeutic target for KRAS-driven colorectal cancers.

7.
Nat Commun ; 11(1): 3701, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32709883

ABSTRACT

Despite its importance in human cancers, including colorectal cancers (CRC), oncogenic KRAS has been extremely challenging to target therapeutically. To identify potential vulnerabilities in KRAS-mutated CRC, we characterize the impact of oncogenic KRAS on the cell surface of intestinal epithelial cells. Here we show that oncogenic KRAS alters the expression of a myriad of cell-surface proteins implicated in diverse biological functions, and identify many potential surface-accessible therapeutic targets. Cell surface-based loss-of-function screens reveal that ATP7A, a copper-exporter upregulated by mutant KRAS, is essential for neoplastic growth. ATP7A is upregulated at the surface of KRAS-mutated CRC, and protects cells from excess copper-ion toxicity. We find that KRAS-mutated cells acquire copper via a non-canonical mechanism involving macropinocytosis, which appears to be required to support their growth. Together, these results indicate that copper bioavailability is a KRAS-selective vulnerability that could be exploited for the treatment of KRAS-mutated neoplasms.


Subject(s)
Colorectal Neoplasms/metabolism , Copper/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Biological Availability , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Copper-Transporting ATPases/metabolism , Female , Humans , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Mice, Nude , Mice, SCID , Mutation
8.
Cell Rep ; 31(7): 107660, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433969

ABSTRACT

In human cells, the expression of ∼1,000 genes is modulated throughout the cell cycle. Although some of these genes are controlled by specific transcriptional programs, very little is known about their post-transcriptional regulation. Here, we analyze the expression signature associated with all 687 RNA-binding proteins (RBPs) and identify 39 that significantly correlate with cell cycle mRNAs. We find that NF45 and NF90 play essential roles in mitosis, and transcriptome analysis reveals that they are necessary for the expression of a subset of mitotic mRNAs. Using proteomics, we identify protein clusters associated with the NF45-NF90 complex, including components of Staufen-mediated mRNA decay (SMD). We show that depletion of SMD components increases the binding of mitotic mRNAs to the NF45-NF90 complex and rescues cells from mitotic defects. Together, our results indicate that the NF45-NF90 complex plays essential roles in mitosis by competing with the SMD machinery for a common set of mRNAs.


Subject(s)
Mitosis/physiology , Nuclear Factor 45 Protein/metabolism , Nuclear Factor 90 Proteins/metabolism , RNA Stability/physiology , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Mitosis/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Factor 45 Protein/genetics , Nuclear Factor 90 Proteins/genetics , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
9.
Mol Cell Proteomics ; 19(1): 50-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31678930

ABSTRACT

The RAS/mitogen-activated protein kinase (MAPK) signaling pathway regulates various biological functions, including cell survival, proliferation and migration. This pathway is frequently deregulated in cancer, including melanoma, which is the most aggressive form of skin cancer. RSK (p90 ribosomal S6 kinase) is a MAPK-activated protein kinase required for melanoma growth and proliferation, but relatively little is known about its function and the nature of its cellular partners. In this study, we used a proximity-based labeling approach to identify RSK proximity partners in cells. We identified many potential RSK-interacting proteins, including p120ctn (p120-catenin), which is an essential component of adherens junction (AJ). We found that RSK phosphorylates p120ctn on Ser320, which appears to be constitutively phosphorylated in melanoma cells. We also found that RSK inhibition increases melanoma cell-cell adhesion, suggesting that constitutive RAS/MAPK signaling negatively regulates AJ integrity. Together, our results indicate that RSK plays an important role in the regulation of melanoma cell-cell adhesion.


Subject(s)
Catenins/metabolism , Cell Adhesion/genetics , Melanoma/metabolism , Proteomics/methods , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Catenins/genetics , Cell Line, Tumor , HEK293 Cells , Humans , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , RNA Interference , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction/genetics , Delta Catenin
10.
Blood Adv ; 3(21): 3307-3321, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31698461

ABSTRACT

Acute megakaryoblastic leukemia (AMKL) represents ∼10% of pediatric acute myeloid leukemia cases and typically affects young children (<3 years of age). It remains plagued with extremely poor treatment outcomes (<40% cure rates), mostly due to primary chemotherapy refractory disease and/or early relapse. Recurrent and mutually exclusive chimeric fusion oncogenes have been detected in 60% to 70% of cases and include nucleoporin 98 (NUP98) gene rearrangements, most commonly NUP98-KDM5A. Human models of NUP98-KDM5A-driven AMKL capable of faithfully recapitulating the disease have been lacking, and patient samples are rare, further limiting biomarkers and drug discovery. To overcome these impediments, we overexpressed NUP98-KDM5A in human cord blood hematopoietic stem and progenitor cells using a lentiviral-based approach to create physiopathologically relevant disease models. The NUP98-KDM5A fusion oncogene was a potent inducer of maturation arrest, sustaining long-term proliferative and progenitor capacities of engineered cells in optimized culture conditions. Adoptive transfer of NUP98-KDM5A-transformed cells into immunodeficient mice led to multiple subtypes of leukemia, including AMKL, that phenocopy human disease phenotypically and molecularly. The integrative molecular characterization of synthetic and patient NUP98-KDM5A AMKL samples revealed SELP, MPIG6B, and NEO1 as distinctive and novel disease biomarkers. Transcriptomic and proteomic analyses pointed to upregulation of the JAK-STAT signaling pathway in the model AMKL. Both synthetic models and patient-derived xenografts of NUP98-rearranged AMKL showed in vitro therapeutic vulnerability to ruxolitinib, a clinically approved JAK2 inhibitor. Overall, synthetic human AMKL models contribute to defining functional dependencies of rare genotypes of high-fatality pediatric leukemia, which lack effective and rationally designed treatments.


Subject(s)
Biomarkers , Disease Models, Animal , Leukemia, Megakaryoblastic, Acute/etiology , Leukemia, Megakaryoblastic, Acute/pathology , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Retinoblastoma-Binding Protein 2/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Computational Biology/methods , Disease Susceptibility , Gene Expression , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunophenotyping , Leukemia, Megakaryoblastic, Acute/therapy , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Retinoblastoma-Binding Protein 2/metabolism , Xenograft Model Antitumor Assays
11.
Cancer Lett ; 449: 196-206, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30771434

ABSTRACT

ProNGF expression has been linked to several types of cancers including breast cancer, and we have previously shown that proNGF stimulates breast cancer invasion in an autocrine manner through membrane receptors sortilin and TrkA. However, little is known regarding TrkA-associated protein partners upon proNGF stimulation. By proteomic analysis and proximity ligation assays, we found that proNGF binding to sortilin induced sequential formation of the functional sortilin/TrkA/EphA2 complex, leading to TrkA-phosphorylation dependent Akt activation and EphA2-dependent Src activation. EphA2 inhibition using siRNA approach abolished proNGF-stimulated clonogenic growth of breast cancer cell lines. Combinatorial targeting of TrkA and EphA2 dramatically reduced colony formation in vitro, primary tumor growth and metastatic dissemination towards the brain in vivo. Finally, proximity ligation assay in breast tumor samples revealed that increased TrkA/EphA2 proximity ligation assay signals were correlated with a decrease of overall survival in patients. All together, these data point out the importance of TrkA/EphA2 functional association in proNGF-induced tumor promoting effects, and provide a rationale to target proNGF/TrkA/EphA2 axis by alternative methods other than the simple use of tyrosine kinase inhibitors in breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Cell Proliferation , Ephrin-A2/metabolism , Nerve Growth Factor/metabolism , Protein Precursors/metabolism , Receptor, trkA/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/enzymology , Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Ephrin-A2/genetics , Female , Humans , MCF-7 Cells , Mice, SCID , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , RNAi Therapeutics , Receptor, EphA2 , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
12.
Cell Rep ; 23(5): 1476-1490, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29719259

ABSTRACT

AXL is activated by its ligand GAS6 and is expressed in triple-negative breast cancer cells. In the current study, we report AXL expression in HER2-positive (HER2+) breast cancers where it correlates with poor patient survival. Using murine models of HER2+ breast cancer, Axl, but not its ligand Gas6, was found to be essential for metastasis. We determined that AXL is required for intravasation, extravasation, and growth at the metastatic site. We found that AXL is expressed in HER2+ cancers displaying epithelial-to-mesenchymal transition (EMT) signatures where it contributes to sustain EMT. Interfering with AXL in a patient-derived xenograft (PDX) impaired transforming growth factor ß (TGF-ß)-induced cell invasion. Last, pharmacological inhibition of AXL specifically decreased the metastatic burden of mice developing HER2+ breast cancer. Our data identify AXL as a potential anti-metastatic co-therapeutic target for the treatment of HER2+ breast cancers.


Subject(s)
Breast Neoplasms/mortality , Epithelial-Mesenchymal Transition , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Heterografts , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor, ErbB-2/genetics , Axl Receptor Tyrosine Kinase
13.
Blood ; 129(25): 3344-3351, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28408459

ABSTRACT

A small subset of human cord blood CD34+ cells express endothelial protein C receptor (EPCR/CD201/PROCR) when exposed to the hematopoietic stem cell (HSC) self-renewal agonist UM171. In this article, we show that EPCR-positive UM171-treated cells, as opposed to EPCR-negative cells, exhibit robust multilineage repopulation and serial reconstitution ability in immunocompromised mice. In contrast to other stem cell markers, such as CD38, EPCR expression is maintained when cells are introduced in culture, irrespective of UM171 treatment. Although engineered overexpression of EPCR fails to reproduce the effects of UM171 on HSC activity, its expression is required for the repopulating activity of human HSCs. Altogether, our results indicate that EPCR is a reliable and cell culture-compatible marker of UM171-expanded human cord blood HSCs.


Subject(s)
Antigens, CD34/analysis , Antigens, CD/analysis , Fetal Blood/cytology , Hematopoietic Stem Cells/drug effects , Indoles/pharmacology , Pyrimidines/pharmacology , Receptors, Cell Surface/analysis , Animals , Cell Culture Techniques , Cell Proliferation/drug effects , Endothelial Protein C Receptor , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Mice, Inbred NOD , Mice, SCID
14.
Mol Cell Biol ; 37(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28096188

ABSTRACT

The scaffolding adapter protein Gab2 (Grb2-associated binder) promotes cell proliferation, survival, and motility by engaging several signaling pathways downstream of growth factor and cytokine receptors. In particular, Gab2 plays essential roles in mast cells, as it is required for phosphoinositide 3-kinase (PI3K) activation in response to Kit and the high-affinity IgE receptor. While the positive role of Gab2 in PI3K signaling is well documented, very little is known about the mechanisms that attenuate its function. Here we show that Gab2 becomes phosphorylated on multiple proline-directed sites upon stimulation of the Ras/extracellular signal-regulated kinase (ERK) signaling pathway. We demonstrate that ERK1 and ERK2 interact with Gab2 via a novel docking motif, which is required for subsequent Gab2 phosphorylation in response to ERK1/2 activation. We identified four ERK1/2-dependent phosphorylation sites in Gab2 that prevent the recruitment of the p85 regulatory subunit of PI3K. Using bone marrow-derived mast cells to study Gab2-dependent signaling, we found that the inhibition of ERK1/2 activity promotes Akt signaling in response to Kit and the high-affinity IgE receptor. Together, our results indicate that ERK1/2 participates in a negative-feedback loop that attenuates PI3K/Akt signaling in response to various agonists.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , GRB2 Adaptor Protein/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Amino Acid Sequence , GRB2 Adaptor Protein/chemistry , HEK293 Cells , Humans , Mast Cells/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Phosphorylation , Protein Domains , ras Proteins/metabolism
15.
Oncotarget ; 6(12): 9807-19, 2015.
Article in English | MEDLINE | ID: mdl-25840418

ABSTRACT

There is accumulating evidence that TrkA and its ligand Nerve Growth Factor (NGF) are involved in cancer development. Staurosporine derivatives such as K252a and lestaurtinib have been developed to block TrkA kinase signaling, but no clinical trial has fully demonstrated their therapeutic efficacy. Therapeutic failures are likely due to the existence of intrinsic signaling pathways in cancer cells that impede or bypass the effects of TrkA tyrosine kinase inhibitors. To verify this hypothesis, we combined different approaches including mass spectrometry proteomics, co-immunoprecipitation and proximity ligation assays. We found that NGF treatment induced CD44 binding to TrkA at the plasma membrane and subsequent activation of the p115RhoGEF/RhoA/ROCK1 pathway to stimulate breast cancer cell invasion. The NGF-induced CD44 signaling was independent of TrkA kinase activity. Moreover, both TrkA tyrosine kinase inhibition with lestaurtinib and CD44 silencing with siRNA inhibited cell growth in vitro as well as tumor development in mouse xenograft model; combined treatment significantly enhanced the antineoplastic effects of either treatment alone. Altogether, our results demonstrate that NGF-induced tyrosine kinase independent TrkA signaling through CD44 was sufficient to maintain tumor aggressiveness. Our findings provide an alternative mechanism of cancer resistance to lestaurtinib and indicate that dual inhibition of CD44 and TrkA tyrosine kinase activity may represent a novel therapeutic strategy.


Subject(s)
Carbazoles/therapeutic use , Drug Resistance, Neoplasm , Hyaluronan Receptors/metabolism , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Animals , Biotinylation , Cell Line, Tumor , Cell Membrane/metabolism , Female , Furans , Gene Silencing , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Binding , Proteomics , RNA, Small Interfering/metabolism , Signal Transduction
16.
Nanomedicine ; 10(8): 1667-77, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24842766

ABSTRACT

In this work, a novel ferrocenyl complex (ansa-FcdiOH) was assessed for brain tumor therapy through stealth lipid nanocapsules (LNCs). Stealth LNCs, prepared according to a one-step process, showed rapid uptake by cancer cells and extended blood circulation time. The ferrocenyl complex was successfully encapsulated into these LNCs measuring 40 nm with a high loading capacity (6.4%). In vitro studies showed a potent anticancer effect of ansa-FcdiOH on 9L cells with a low IC50 value (0.1 µM) associated with an oxidative stress and a dose-dependent alteration of the cell cycle. Repeated intravenous injections of stealth ansa-FcdiOH LNCs in ectopic glioma bearing rats induced a significant tumor growth inhibition, supported by a reduced number of proliferative cells in tumors compared to control group. Additionally, no liver damage was observed in treated animals. These results indicated that stealth ansa-FcdiOH LNCs might be considered as a potential new approach for cancer chemotherapy. FROM THE CLINICAL EDITOR: In this study, a novel ferrocenyl complex was assessed for brain tumor therapy through stealth lipid nanocapsules, demonstrating no liver damage, and superior tumor volume reduction compared to saline and stealth lipid nanocapsules alone in an ectopic glioma model.


Subject(s)
Ferrous Compounds/chemistry , Ferrous Compounds/therapeutic use , Glioma/drug therapy , Nanocapsules/chemistry , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Drug Carriers/chemistry , Female , Nanomedicine , Rats , Rats, Inbred F344 , Reactive Oxygen Species
SELECTION OF CITATIONS
SEARCH DETAIL