Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 92
Filtrer
1.
Mol Carcinog ; 63(7): 1248-1259, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38558423

RÉSUMÉ

Epithelial ovarian cancers that are nonhomologous recombination deficient, as well as those that are recurrent and in a platinum-resistant state, have limited therapeutic options. The objectives of this study were to characterize the mechanism of action and investigate the therapeutic potential of a small molecule, VDX-111, against ovarian cancer. We examined the ability of VDX-111 to inhibit the growth of a panel of ovarian cancer cell lines, focusing on BRCA wild-type lines. We found that VDX-111 causes a dose-dependent loss of cell viability across ovarian cancer cell lines. Reverse phase protein array (RPPA) analysis was used to identify changes in cell signaling in response to VDX-111 treatment. An RPPA analysis performed on cells treated with VDX-111 detected changes in cell signaling related to autophagy and necroptosis. Immunoblots of OVCAR3 and SNU8 cells confirmed a dose-dependent increase in LC3A/B and RIPK1. Incucyte live cell imaging was used to measure cell proliferation and death in response to VDX-111 alone and with inhibitors of apoptosis, necroptosis, and autophagy. Annexin/PI assays suggested predominantly nonapoptotic cell death, while real-time kinetic imaging of cell growth indicated the necroptosis inhibitor, necrostatin-1, attenuates VDX-111-induced loss of cell viability, suggesting a necroptosis-dependent mechanism. Furthermore, VDX-111 inhibited tumor growth in patient-derived xenograft and syngeneic murine models. In conclusion, the cytotoxic effects of VDX-111 seen in vitro and in vivo appear to occur in a necroptosis-dependent manner and may promote an antitumor immune response.


Sujet(s)
Prolifération cellulaire , Nécroptose , Tumeurs de l'ovaire , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Femelle , Animaux , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Nécroptose/effets des médicaments et des substances chimiques , Souris , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Évolution de la maladie , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Carcinome épithélial de l'ovaire/traitement médicamenteux , Carcinome épithélial de l'ovaire/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Imidazoles/pharmacologie
2.
Cancers (Basel) ; 16(8)2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38672531

RÉSUMÉ

The addition of the proteasome inhibitor bortezomib to standard chemotherapy did not improve survival in pediatric acute myeloid leukemia (AML) when all patients were analyzed as a group in the Children's Oncology Group phase 3 trial AAML1031 (NCT01371981). Proteasome inhibition influences the chromatin landscape and proteostasis, and we hypothesized that baseline proteomic analysis of histone- and chromatin-modifying enzymes (HMEs) would identify AML subgroups that benefitted from bortezomib addition. A proteomic profile of 483 patients treated with AAML1031 chemotherapy was generated using a reverse-phase protein array. A relatively high expression of 16 HME was associated with lower EFS and higher 3-year relapse risk after AML standard treatment compared to low expressions (52% vs. 29%, p = 0.005). The high-HME profile correlated with more transposase-accessible chromatin, as demonstrated via ATAC-sequencing, and the bortezomib addition improved the 3-year overall survival compared with standard therapy (62% vs. 75%, p = 0.033). These data suggest that there are pediatric AML populations that respond well to bortezomib-containing chemotherapy.

3.
Cancer Sci ; 115(5): 1378-1387, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38409909

RÉSUMÉ

The last few decades have seen remarkable strides in the field of cancer therapy. Precision oncology coupled with comprehensive genomic profiling has become routine clinical practice for solid tumors, the advent of immune checkpoint inhibitors has transformed the landscape of oncology treatment, and the number of cancer drug approvals has continued to increase. Nevertheless, the application of genomics-driven precision oncology has thus far benefited only 10%-20% of cancer patients, leaving the majority without matched treatment options. This limitation underscores the need to explore alternative avenues with regard to selecting patients for targeted therapies. In contrast with genomics-based approaches, proteomics-based strategies offer a more precise understanding of the intricate biological processes driving cancer pathogenesis. This perspective underscores the importance of integrating complementary proteomic analyses into the next phase of precision oncology to establish robust biomarker-drug associations and surmount challenges related to drug resistance. One promising technology in this regard is the reverse-phase protein array (RPPA), which excels in quantitatively detecting protein modifications, even with limited amounts of sample. Its cost-effectiveness and rapid turnaround time further bolster its appeal for application in clinical settings. Here, we review the current status of genomics-driven precision oncology, as well as its limitations, with an emphasis on drug resistance. Subsequently, we explore the application of RPPA technology as a catalyst for advancing precision oncology. Through illustrative examples drawn from clinical trials, we demonstrate its utility for unraveling the molecular mechanisms underlying drug responses and resistance.


Sujet(s)
Tumeurs , Médecine de précision , Analyse par réseau de protéines , Protéomique , Humains , Médecine de précision/méthodes , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Analyse par réseau de protéines/méthodes , Protéomique/méthodes , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Génomique/méthodes , Oncologie médicale/méthodes , Résistance aux médicaments antinéoplasiques , Thérapie moléculaire ciblée/méthodes
4.
CNS Neurosci Ther ; 30(2): e14407, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-37564024

RÉSUMÉ

AIMS: This study aims to reveal a promising biomarker for Parkinson's disease (PD) based on research with reverse phase protein array (RPPA) technology for the first time and in vivo verification, which gains time for early intervention in PD, thus increasing the effectiveness of treatment and reducing disease morbidity. METHODS AND RESULTS: We employed RPPA technology which can assess both total and post-translationally modified proteins to identify biomarker candidates of PD in a cellular PD model. As a result, the phosphorylation (pY-1248) of the epidermal growth factor receptor (EGFR) ErbB2 is a promising biomarker candidate for PD. In addition, lapatinib, an ErbB2 tyrosine kinase inhibitor, was used to verify this PD biomarker candidate in vivo. We found that lapatinib-attenuated dopaminergic neuron loss and PD-like behavior in the zebrafish PD model. Accordingly, the expression of ErbB2pY-1248 significantly increased in the MPTP-induced mouse PD model. Our results suggest that ErbB2pY-1248 is a predictive biomarker for PD. CONCLUSIONS: In this study, we found that ErbB2pY-1248 is a predictive biomarker of PD by using RPPA technology and in vivo verification. It offers a new perspective on PD diagnosing and treatment, which will be essential in identifying individuals at risk of PD. In addition, this study provides new ideas for digging into biomarkers of other neurodegenerative diseases.


Sujet(s)
Maladie de Parkinson , Animaux , Souris , Maladie de Parkinson/diagnostic , Maladie de Parkinson/métabolisme , Danio zébré , Lapatinib/métabolisme , Dopamine/métabolisme , Neurones dopaminergiques/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine
5.
Front Aging Neurosci ; 15: 1060186, 2023.
Article de Anglais | MEDLINE | ID: mdl-37261265

RÉSUMÉ

Introduction: The development and maintenance of neural circuits is highly sensitive to neural activity. General anesthetics have profound effects on neural activity and, as such, there is concern that these agents may alter cellular integrity and interfere with brain wiring, such as when exposure occurs during the vulnerable period of brain development. Under those conditions, exposure to anesthetics in clinical use today causes changes in synaptic strength and number, widespread apoptosis, and long-lasting cognitive impairment in a variety of animal models. Remarkably, most anesthetics produce these effects despite having differing receptor mechanisms of action. We hypothesized that anesthetic agents mediate these effects by inducing a shared signaling pathway. Methods: We exposed cultured cortical cells to propofol, etomidate, or dexmedetomidine and assessed the protein levels of dozens of signaling molecules and post-translational modifications using reverse phase protein arrays. To probe the role of neural activity, we performed separate control experiments to alter neural activity with non-anesthetics. Having identified anesthetic-induced changes in vitro, we investigated expression of the target proteins in the cortex of sevoflurane anesthetized postnatal day 7 mice by Western blotting. Results: All the anesthetic agents tested in vitro reduced phosphorylation of the ribosomal protein S6, an important member of the mTOR signaling pathway. We found a comparable decrease in cortical S6 phosphorylation by Western blotting in sevoflurane anesthetized neonatal mice. Using a systems approach, we determined that propofol, etomidate, dexmedetomidine, and APV/TTX all similarly modulate a signaling module that includes pS6 and other cell mediators of the mTOR-signaling pathway. Discussion: Reduction in S6 phosphorylation and subsequent suppression of the mTOR pathway may be a common and novel signaling event that mediates the impact of general anesthetics on neural circuit development.

6.
J Neurooncol ; 163(2): 327-338, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37237151

RÉSUMÉ

BACKGROUND: Glioblastoma (GBM) is an aggressive brain cancer that typically results in death in the first 15 months after diagnosis. There have been limited advances in finding new treatments for GBM. In this study, we investigated molecular differences between patients with extremely short (≤ 9 months, Short term survivors, STS) and long survival (≥ 36 months, Long term survivors, LTS). METHODS: Patients were selected from an in-house cohort (GLIOTRAIN-cohort), using defined inclusion criteria (Karnofsky score > 70; age < 70 years old; Stupp protocol as first line treatment, IDH wild type), and a multi-omic analysis of LTS and STS GBM samples was performed. RESULTS: Transcriptomic analysis of tumour samples identified cilium gene signatures as enriched in LTS. Moreover, Immunohistochemical analysis confirmed the presence of cilia in the tumours of LTS. Notably, reverse phase protein array analysis (RPPA) demonstrated increased phosphorylated GAB1 (Y627), SRC (Y527), BCL2 (S70) and RAF (S338) protein expression in STS compared to LTS. Next, we identified 25 unique master regulators (MR) and 13 transcription factors (TFs) belonging to ontologies of integrin signalling and cell cycle to be upregulated in STS. CONCLUSION: Overall, comparison of STS and LTS GBM patients, identifies novel biomarkers and potential actionable therapeutic targets for the management of GBM.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Humains , Sujet âgé , Glioblastome/anatomopathologie , Pronostic , Tumeurs du cerveau/anatomopathologie , Encéphale/anatomopathologie , Survivants
7.
J Pers Med ; 12(8)2022 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-36013226

RÉSUMÉ

Triple negative breast cancer (TNBC) remains a therapeutic challenge due to the lack of targetable genetic alterations and the frequent development of resistance to the standard cisplatin-based chemotherapies. Here, we have taken a systems biology approach to investigate kinase signal transduction networks that are involved in TNBC resistance to cisplatin. Treating a panel of cisplatin-sensitive and cisplatin-resistant TNBC cell lines with a panel of kinase inhibitors allowed us to reconstruct two kinase signalling networks that characterise sensitive and resistant cells. The analysis of these networks suggested that the activation of the PI3K/AKT signalling pathway is critical for cisplatin resistance. Experimental validation of the computational model predictions confirmed that TNBC cell lines with activated PI3K/AKT signalling are sensitive to combinations of cisplatin and PI3K/AKT pathway inhibitors. Thus, our results reveal a new therapeutic approach that is based on identifying targeted therapies that synergise with conventional chemotherapies.

8.
Expert Rev Proteomics ; 19(2): 115-129, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35466854

RÉSUMÉ

INTRODUCTION: Drug resistance is the main barrier to achieving cancer cures with medical therapy. Cancer drug resistance occurs, in part, due to adaptation of the tumor and microenvironment to therapeutic stress at a proteomic level. Reverse-phase protein arrays (RPPA) are well suited to proteomic analysis of drug resistance due to high sample throughput, sensitive detection of phosphoproteins, and validation for a large number of critical cellular pathways. AREAS COVERED: This review summarizes contributions of RPPA to understanding and combating drug resistance. In particular, contributions of RPPA to understanding resistance to PARP inhibitors, BRAF inhibitors, immune checkpoint inhibitors, and breast cancer investigational therapies are discussed. Articles reviewed were identified by MEDLINE, Scopus, and Cochrane search for keywords 'proteomics,' 'reverse-phase protein array,' 'drug resistance,' 'PARP inhibitor,' 'BRAF inhibitor,' 'immune checkpoint inhibitor,' and 'I-SPY' spanning October 1, 1960 - October 1, 2021. EXPERT OPINION: Precision oncology has thus far failed to convert the armament of targeted therapies into durable responses for most patients, highlighting that genetic sequencing alone is insufficient to guide therapy selection and overcome drug resistance. Combined genomic and proteomic analyses paired with creative drug combinations and dosing strategies hold promise for maturing precision oncology into an era of improved patient outcomes.


Sujet(s)
Antinéoplasiques , Tumeurs du sein , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Tumeurs du sein/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Humains , Médecine de précision , Analyse par réseau de protéines , Inhibiteurs de protéines kinases , Protéomique , Protéines proto-oncogènes B-raf , Microenvironnement tumoral
9.
Nutrients ; 14(5)2022 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-35267943

RÉSUMÉ

Esophageal adenocarcinoma (EAC) is a cancer characterized by rapidly rising incidence and poor survival, resulting in the need for new prevention and treatment options. We utilized two cranberry polyphenol extracts, one proanthocyanidin enriched (C-PAC) and a combination of anthocyanins, flavonoids, and glycosides (AFG) to assess inhibitory mechanisms utilizing premalignant Barrett's esophagus (BE) and EAC derived cell lines. We employed reverse phase protein arrays (RPPA) and Western blots to examine cancer-associated pathways and specific signaling cascades modulated by C-PAC or AFG. Viability results show that C-PAC is more potent than AFG at inducing cell death in BE and EAC cell lines. Based on the RPPA results, C-PAC significantly modulated 37 and 69 proteins in JH-EsoAd1 (JHAD1) and OE19 EAC cells, respectively. AFG treatment significantly altered 49 proteins in both JHAD1 and OE19 cells. Bioinformatic analysis of RPPA results revealed many previously unidentified pathways as modulated by cranberry polyphenols including NOTCH signaling, immune response, and epithelial to mesenchymal transition. Collectively, these results provide new insight regarding mechanisms by which cranberry polyphenols exert cancer inhibitory effects targeting EAC, with implications for potential use of cranberry constituents as cancer preventive agents.


Sujet(s)
Tumeurs de l'oesophage , Vaccinium macrocarpon , Anthocyanes/pharmacologie , Transition épithélio-mésenchymateuse , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/prévention et contrôle , Extraits de plantes/pharmacologie , Polyphénols/pharmacologie
10.
Mol Oncol ; 16(5): 1072-1090, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34856074

RÉSUMÉ

A more comprehensive understanding of how cells respond to drug intervention, the likely immediate signalling responses and how resistance may develop within different microenvironments will help inform treatment regimes. The nonreceptor tyrosine kinase SRC regulates many cellular signalling processes, and pharmacological inhibition has long been a target of cancer drug discovery projects. Here, we describe the in vitro and in vivo characterisation of the small-molecule SRC inhibitor AZD0424. We show that AZD0424 potently inhibits the phosphorylation of tyrosine-419 of SRC (IC50 ~ 100 nm) in many cancer cell lines; however, inhibition of cell viability, via a G1 cell cycle arrest, was observed only in a subset of cancer cell lines in the low (on target) micromolar range. We profiled the changes in intracellular pathway signalling in cancer cells following exposure to AZD0424 and other targeted therapies using reverse-phase protein array (RPPA) analysis. We demonstrate that SRC is activated in response to treatment of KRAS-mutant colorectal cell lines with MEK inhibitors (trametinib or AZD6244) and that AZD0424 abrogates this. Cell lines treated with trametinib or AZD6244 in combination with AZD0424 had reduced EGFR, FAK and SRC compensatory activation, and cell viability was synergistically inhibited. In vivo, trametinib treatment of mice-bearing HCT116 tumours increased phosphorylation of SRC on Tyr419, and, when combined with AZD0424, inhibition of tumour growth was greater than with trametinib alone. We also demonstrate that drug-induced resistance to trametinib is not re-sensitised by AZD0424 treatment in vitro, likely as a result of multiple compensatory signalling mechanisms; however, inhibition of SRC remains an effective way to block invasion of trametinib-resistant tumour cells. These data imply that SRC inhibition may offer a useful addition to MEK inhibitor combination strategies.


Sujet(s)
Tumeurs , Quinazolines , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Humains , Souris , Mitogen-Activated Protein Kinase Kinases/métabolisme , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Quinazolines/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Cells ; 10(10)2021 10 15.
Article de Anglais | MEDLINE | ID: mdl-34685748

RÉSUMÉ

Triple-negative breast cancer (TNBC) is a subtype of breast cancer that comprises various disease entities, all of which share a set of common features: a lack of expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2, respectively. Because of their receptor status, conventional chemotherapy remains the main therapeutic option for TNBC patients. We employed a reverse phase protein array approach (RPPA), complemented by immunohistochemistry, to quantitatively profile the activation state of 84 actionable key signaling intermediates and phosphoproteins in a set of 44 TNBC samples. We performed supervised and unsupervised approaches to proteomic data analysis to identify groups of samples sharing common characteristics that could be amenable to existing therapies. We found the heterogenous activation of multiple pathways, with PI3 K/AKT/mTOR signaling being the most common event. Some specific individualized therapeutic possibilities include the expression of oncogenic KIT in association with cytokeratin 15 and Erk1/2 positive tumors, both of which may have clinical value.


Sujet(s)
Protéomique , Tumeurs du sein triple-négatives/métabolisme , Carcinogenèse/anatomopathologie , Humains , Protéines tumorales/métabolisme , Phosphorylation , Analyse par réseau de protéines , Protéines proto-oncogènes c-kit/génétique , Transduction du signal
12.
Int J Mol Sci ; 22(16)2021 Aug 11.
Article de Anglais | MEDLINE | ID: mdl-34445353

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the third leading cause of cancer death worldwide. Additionally, the efficacy of targeted molecular therapies with multiple tyrosine kinase inhibitors is limited. In this study, we focused on the cellular signaling pathways common to diverse HCC cells and used quantitative reverse phase protein array (RPPA) and statistical analyses to elucidate the molecular mechanisms determining its malignancy. We examined the heterogeneity of 17 liver cancer cell lines by performing cluster analysis of their expression of CD90 and EpCAM cancer stem cell markers. Gaussian mixture model clustering identified three dominant clusters: CD90-positive and EpCAM-negative (CD90+), EpCAM-positive and CD90-negative (EpCAM+) and EpCAM-negative and CD90-negative (Neutral). A multivariate analysis by partial least squares revealed that the former two cell populations showed distinct patterns of protein expression and phosphorylation in the EGFR and EphA2 signaling pathways. The CD90+ cells exhibited higher abundance of AKT, EphA2 and its phosphorylated form at Ser897, whereas the EpCAM+ cells exhibited higher abundance of ERK, RSK and its phosphorylated form. This demonstrates that pro-oncogenic, ligand-independent EphA2 signaling plays a dominant role in CD90+ cells with higher motility and metastatic activity than EpCAM+ cells. We also showed that an AKT inhibitor reduced the proliferation and survival of CD90+ cells but did not affect those of EpCAM+ cells. Taken together, our results suggest that AKT activation may be a key pro-oncogenic regulator in HCC.


Sujet(s)
Carcinome hépatocellulaire/anatomopathologie , Molécule d'adhérence des cellules épithéliales/métabolisme , Tumeurs du foie/anatomopathologie , Récepteur EphA2/physiologie , Antigènes Thy-1/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Lignée cellulaire tumorale , Cellules HepG2 , Humains , Tumeurs du foie/métabolisme , Cellules souches tumorales/métabolisme , Récepteur EphA2/métabolisme , Transduction du signal
13.
Front Neurol ; 12: 654606, 2021.
Article de Anglais | MEDLINE | ID: mdl-34122302

RÉSUMÉ

Epilepsy is characterized by recurrent, spontaneous seizures and is a major contributor to the global burden of neurological disease. Although epilepsy can result from a variety of brain insults, in many cases the cause is unknown and, in a significant proportion of cases, seizures cannot be controlled by available treatments. Understanding the molecular alterations that underlie or are triggered by epileptogenesis would help to identify therapeutics to prevent or control progression to epilepsy. To this end, the moderate throughput technique of Reverse Phase Protein Arrays (RPPA) was used to profile changes in protein expression in a pilocarpine mouse model of acquired epilepsy. Levels of 54 proteins, comprising phosphorylation-dependent and phosphorylation-independent components of major signaling pathways and cellular complexes, were measured in hippocampus, cortex and cerebellum of mice at six time points, spanning 15 min to 2 weeks after induction of status epilepticus. Results illustrate the time dependence of levels of the commonly studied MTOR pathway component, pS6, and show, for the first time, detailed responses during epileptogenesis of multiple components of the MTOR, MAPK, JAK/STAT and apoptosis pathways, NMDA receptors, and additional cellular complexes. Also noted are time- and brain region- specific changes in correlations among levels of functionally related proteins affecting both neurons and glia. While hippocampus and cortex are primary areas studied in pilocarpine-induced epilepsy, cerebellum also shows significant time-dependent molecular responses.

14.
Mol Cell Proteomics ; 20: 100094, 2021.
Article de Anglais | MEDLINE | ID: mdl-33991687

RÉSUMÉ

Identifying biomarkers is important for assessment of disease progression, prediction of symptom development, and determination of treatment effectiveness. While unbiased analyses of differential gene expression using next-generation sequencing methods are now routinely conducted, proteomics studies are more challenging because of traditional methods predominantly being low throughput and offering a limited dynamic range for simultaneous detection of hundreds of proteins that drastically differ in their intracellular abundance. We utilized a sensitive and high-throughput proteomic technique, reverse phase protein array (RPPA), to attain protein expression profiles of primary fibroblasts obtained from patients with Friedreich's ataxia (FRDA) and unaffected controls (CTRLs). The RPPA was designed to detect 217 proteins or phosphorylated proteins by individual antibody, and the specificity of each antibody was validated prior to the experiment. Among 62 fibroblast samples (44 FRDA and 18 CTRLs) analyzed, 30 proteins/phosphoproteins were significantly changed in FRDA fibroblasts compared with CTRL cells (p < 0.05), mostly representing signaling molecules and metabolic enzymes. As expected, frataxin was significantly downregulated in FRDA samples, thus serving as an internal CTRL for assay integrity. Extensive bioinformatics analyses were conducted to correlate differentially expressed proteins with critical disease parameters (e.g., selected symptoms, age of onset, guanine-adenine-adenine sizes, frataxin levels, and Functional Assessment Rating Scale scores). Members of the integrin family of proteins specifically associated with hearing loss in FRDA. Also, RPPA data, combined with results of transcriptome profiling, uncovered defects in the retinoic acid metabolism pathway in FRDA samples. Moreover, expression of aldehyde dehydrogenase family 1 member A3 differed significantly between cardiomyopathy-positive and cardiomyopathy-negative FRDA cohorts, demonstrating that metabolites such as retinol, retinal, or retinoic acid could become potential predictive biomarkers of cardiac presentation in FRDA.


Sujet(s)
Cardiomyopathies/métabolisme , Ataxie de Friedreich/métabolisme , Rétinoïdes/métabolisme , Adolescent , Adulte , Sujet âgé , Aldehyde oxidoreductases/métabolisme , Marqueurs biologiques/métabolisme , Cellules cultivées , Femelle , Fibroblastes/métabolisme , Humains , Protéines de liaison au fer/métabolisme , Mâle , Adulte d'âge moyen , Analyse par réseau de protéines , Protéomique , Jeune adulte ,
15.
Invest New Drugs ; 39(1): 163-174, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32737717

RÉSUMÉ

BACKGROUND: This Phase Ib study explored combination dosing of the allosteric MEK1/2 inhibitor cobimetinib and the ATP-competitive pan-AKT inhibitor ipatasertib. METHODS: Patients with advanced solid tumors were enrolled to two dose escalation arms, each using a 3 + 3 design in 28-day cycles. In Arm A, patients received concurrent cobimetinib and ipatasertib on days 1-21. In Arm B, cobimetinib was administered intermittently with ipatasertib for 21 days. Primary objectives evaluated dose-limiting toxicities (DLTs), maximum tolerated doses (MTD), and the recommended Phase II dose (RP2D). Secondary objectives included analysis of pharmacokinetic parameters, MAPK and PI3K pathway alterations, changes in tissue biomarkers, and preliminary anti-tumor efficacy. Expansion cohorts included patients with PTEN-deficient triple-negative breast cancer and endometrial cancer. RESULTS: Among 66 patients who received ≥1 dose of study drug, all experienced an adverse event (AE). Although no DLTs were reported, 6 patients experienced Cycle 1 DLT-equivalent AEs. The most common treatment-related AEs were diarrhea, nausea, vomiting, dermatitis acneiform, and fatigue. Thirty-five (53%) patients experienced drug-related AEs of ≥ grade 3 severity. Cobimetinb/ipatasertib MTDs were 60/200 mg on Arm A and 150/300 mg on Arm B; the latter was chosen as the RP2D. No pharmacokinetic interactions were identified. Biomarker analyses indicated pathway blockade and increases in IFNγ and PD-L1 gene expression following the combination. Three patients with endometrial or ovarian cancer achieved partial response, all with PTEN-low disease and two with tumor also harboring KRAS mutation. CONCLUSION: There was limited tolerability and efficacy for this MEK and AKT inhibitor combination. Nonetheless, pharmacodynamic analyses indicated target engagement and suggest rationale for further exploration of cobimetinib or ipatasertib in combination with other anticancer agents. ClinicalTrials.gov identifier: NCT01562275.


Sujet(s)
Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Azétidines/pharmacologie , Azétidines/usage thérapeutique , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Sujet âgé , Sujet âgé de 80 ans ou plus , Antinéoplasiques/effets indésirables , Antinéoplasiques/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Azétidines/effets indésirables , Azétidines/pharmacocinétique , Relation dose-effet des médicaments , Femelle , Humains , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Mitogen-Activated Protein Kinases/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Phosphatidylinositol 3-kinases/effets des médicaments et des substances chimiques , Pipérazines/effets indésirables , Pipérazines/pharmacocinétique , Pipéridines/effets indésirables , Pipéridines/pharmacocinétique , Pyrimidines/effets indésirables , Pyrimidines/pharmacocinétique
16.
Methods Mol Biol ; 2237: 103-122, 2021.
Article de Anglais | MEDLINE | ID: mdl-33237412

RÉSUMÉ

Reverse phase protein arrays (RPPA) are used to quantify proteins and protein posttranslational modifications in cellular lysates and body fluids. RPPA technology is suitable for biomarker discovery, protein pathway profiling, functional phenotype analysis, and drug discovery mechanism of action. The principles of RPPA technology are (a) immobilizing protein-containing specimens on a coated slide in discrete spots, (b) antibody recognition of proteins, (c) amplification chemistries to detect the protein-antibody complex, and (d) quantifying spot intensity. Construction of a RPPA begins with the robotic liquid transfer of protein-containing specimens from microtiter plates onto nitrocellulose-coated slides. The robotic arrayer deposits each sample as discrete spots in an array format. Specimens, controls, and calibrators are printed on each array, thus providing a complete calibrated assay on a single slide. Each RPPA slide is subsequently probed with catalyzed signal amplification chemistries and a single primary antibody, a secondary antibody, and either fluorescent or colorimetric dyes. The focus of this chapter is to describe RPPA detection and imaging using a colorimetric (diaminobenzidine (DAB)) detection strategy.


Sujet(s)
Analyse par réseau de protéines/méthodes , Biphényle-3,3',4,4'-tétraamine/composition chimique , Animaux , Anticorps/immunologie , Lignée cellulaire , Colorimétrie/méthodes , Humains , Dosage immunologique/méthodes , Maturation post-traductionnelle des protéines , Protéome/immunologie , Protéome/métabolisme
17.
Methods Mol Biol ; 2237: 207-215, 2021.
Article de Anglais | MEDLINE | ID: mdl-33237420

RÉSUMÉ

Reverse phase protein array (RPPA), a high-throughput, parallel immunoassay in a dot-blot format, is a powerful tool to quantitatively profile protein expression in multiple samples simultaneously using small amounts of material. Despite its success, analysis of post-translationally modified (PTM) proteins has been limited in RPPA assays, primarily due to relatively low availability of antibodies specific to proteins of PTMs, e.g., glycosylation. Moreover, the high matrix complexity, with tens of thousands of proteins in cell lysates or tissue extracts and the low abundance of proteins with PTMs, makes it extremely challenging to detect these proteins with PTMs. Therefore, there is an urgent need to fill this gap, which would greatly contribute to the analysis of a specific PTM by RPPA. In this chapter, we introduce a novel RPPA platform, termed polymer-based reverse phase glycoprotein array (polyGPA), to measure the variation of glycosylation patterns on a three-dimensionally functionalized RPPA. Without the need of specific antibody towards glycosylation, polyGPA represents a highly sensitive strategy to analyze protein glycosylation in multiple complex biological samples in parallel.


Sujet(s)
Glycoprotéines/sang , Immunotransfert/méthodes , Analyse par réseau de protéines/méthodes , Animaux , Glycoprotéines/immunologie , Glycoprotéines/urine , Glycosylation , Humains , Maturation post-traductionnelle des protéines , Protéome/métabolisme
18.
J Pathol ; 252(3): 317-329, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32737994

RÉSUMÉ

Efficacious therapeutic approaches are urgently needed to improve outcomes in patients with oesophageal adenocarcinoma (OAC). However, oncogenic drivers amenable to targeted therapy are limited and their functional characterisation is essential. Among few targeted therapies available, anti-human epidermal growth factor receptor 2 (HER2) therapy showed only modest benefit for patients with OAC. Herein, we investigated the potential oncogenic role of growth factor receptor bound protein 7 (GRB7), which is reported to be co-amplified with HER2 (ERBB2) in OAC. GRB7 was highly expressed in 15% of OAC tumours, not all of which could be explained by co-amplification with HER2, and was associated with a trend for poorer overall survival. Knockdown of GRB7 decreased proliferation and clonogenic survival, and induced apoptosis. Reverse phase protein array (RPPA) analyses revealed a role for PI3K, mammalian target of rapamycin (mTOR), MAPK, and receptor tyrosine kinase signalling in the oncogenic action of GRB7. Furthermore, the GRB7 and HER2 high-expressing OAC cell line Eso26 showed reduced cell proliferation upon GRB7 knockdown but was insensitive to HER2 inhibition by trastuzumab. Consistent with this, GRB7 knockdown in vivo with an inducible shRNA significantly inhibited tumour growth in cell line xenografts. HER2 expression did not predict sensitivity to trastuzumab, with Eso26 xenografts remaining refractory to trastuzumab treatment. Taken together, our study provides strong evidence for an oncogenic role for GRB7 in OAC and suggests that targeting GRB7 may be a potential therapeutic strategy for this cancer. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Sujet(s)
Adénocarcinome/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Tumeurs de l'oesophage/métabolisme , Protéine adaptatrice GRB7/métabolisme , Adénocarcinome/traitement médicamenteux , Adénocarcinome/mortalité , Adénocarcinome/anatomopathologie , Animaux , Antinéoplasiques immunologiques/usage thérapeutique , Technique de Western , Lignée cellulaire tumorale , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de l'oesophage/mortalité , Tumeurs de l'oesophage/anatomopathologie , Femelle , Techniques de knock-down de gènes , Humains , Immunohistochimie , Souris , Souris de lignée NOD , Souris SCID , Transplantation tumorale , Pronostic , Récepteur ErbB-2/métabolisme , Analyse de survie , Trastuzumab/usage thérapeutique
19.
Leuk Lymphoma ; 61(13): 3052-3065, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-32799592

RÉSUMÉ

Myeloproliferative neoplasms polycythemia vera (PV), essential thrombocythaemia (ET) and primary myelofibrosis constitute a group of haematological diseases. The comprehensive assessment of signaling pathway activation in blood cells may aid the understanding of MPN pathophysiology. Thus, levels of post-translational protein modifications and total protein expression were determined in MPN patients and control leukocytes by using reverse-phase protein arrays (RPPA). Compared to control samples, p-SRC, p-CTNNB1, c-MYC, MCL-1, p-MDM2, BAX and CCNB1 showed higher expression in PV samples than controls. P-JAK2/JAK2 and pro-apoptotic BIM showed differential expression between JAK2V617F-positive and -negative ET patients. Apoptosis, cancer and PI3K/AKT pathways proteins showed differential expression among the studied groups. For most of the proteins analyzed using Western-Blot and RPPA, RPPA showed higher sensitivity to detect subtle differences. Taken together, our data indicate deregulated protein expression in MPN patients compared to controls. Thus, RPPA may be a useful method for broad proteome analysis in MPN patients´ leukocytes.


Sujet(s)
Syndromes myéloprolifératifs , Tumeurs , Humains , Kinase Janus-2/génétique , Mutation , Syndromes myéloprolifératifs/diagnostic , Syndromes myéloprolifératifs/génétique , Phosphatidylinositol 3-kinases , Analyse par réseau de protéines , Protéomique
20.
Int J Cancer ; 147(10): 2891-2901, 2020 11 15.
Article de Anglais | MEDLINE | ID: mdl-32700762

RÉSUMÉ

Antibodies targeting the human epidermal growth factor receptor (EGFR) are used for the treatment of RAS wild-type metastatic colorectal cancer. A significant proportion of patients remains unresponsive to this therapy. Here, we performed a reverse-phase protein array-based (phospho)protein analysis of 63 KRAS, NRAS, BRAF and PIK3CA wild-type metastatic CRC tumours. Responses of tumours to anti-EGFR therapy with cetuximab were recorded in patient-derived xenograft (PDX) models. Unsupervised hierarchical clustering of pretreatment tumour tissue identified three clusters, of which Cluster C3 was exclusively composed of responders. Clusters C1 and C2 exhibited mixed responses. None of the three protein clusters exhibited a significant correlation with transcriptome-based subtypes. Analysis of protein signatures across all PDXs identified 14 markers that discriminated cetuximab-sensitive and cetuximab-resistant tumours: PDK1 (S241), caspase-8, Shc (Y317), Stat3 (Y705), p27, GSK-3ß (S9), HER3, PKC-α (S657), EGFR (Y1068), Akt (S473), S6 ribosomal protein (S240/244), HER3 (Y1289), NF-κB-p65 (S536) and Gab-1 (Y627). Least absolute shrinkage and selection operator and binominal logistic regression analysis delivered refined protein signatures for predicting response to cetuximab. (Phospo-)protein analysis of matched pretreated and posttreated models furthermore showed significant reduction of Gab-1 (Y627) and GSK-3ß (S9) exclusively in responding models, suggesting novel targets for treatment.


Sujet(s)
Cétuximab/administration et posologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/secondaire , Phosphoprotéines/métabolisme , Protéomique/méthodes , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cétuximab/pharmacologie , Phosphatidylinositol 3-kinases de classe I/génétique , Analyse de regroupements , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Femelle , dGTPases/génétique , Humains , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Mâle , Protéines membranaires/génétique , Souris , Phosphoprotéines/effets des médicaments et des substances chimiques , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes p21(ras)/génétique , Apprentissage machine non supervisé , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE