Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
1.
Cancer Lett ; 467: 9-18, 2019 12 28.
Article de Anglais | MEDLINE | ID: mdl-31563561

RÉSUMÉ

Radiation sensitizers that can selectively act on cancer cells hold great promise to patients who receive radiation therapy. We developed a novel targeted therapy and radiation sensitizer for non-small cell lung cancer (NSCLC) based on cetuximab conjugated nanoparticle that targets epidermal growth factor receptor (EGFR) and delivers small interfering RNA (siRNA) against polo-like kinase 1 (PLK1). EGFR is overexpressed in 50% of lung cancer patients and a mediator of DNA repair, while PLK1 is a key mitotic regulator whose inhibition enhances radiation sensitivity. The nanoparticle construct (C-siPLK1-NP) effectively targets EGFR + NSCLC cells and reduces PLK1 expression, leading to G2/M arrest and cell death. Furthermore, we show a synergistic combination between C-siPLK1-NP and radiation, which was confirmed in vivo in A549 flank tumors. We also demonstrate the translational potential of C-siPLK1-NP as a systemic therapeutic in an orthotopic lung tumor model, where administration of C-siPLK1-NP reduced tumor growth and led to prolonged survival. Our findings demonstrate that C-siPLK1-NP is effective as a targeted therapy and as a potent radiation sensitizer for NSCLC. Potential application to other EGFR + cancer types such as colorectal and breast cancer is also demonstrated.


Sujet(s)
Carcinome pulmonaire non à petites cellules/thérapie , Cétuximab/administration et posologie , Tumeurs du poumon/thérapie , Petit ARN interférent/administration et posologie , Radiosensibilisants/administration et posologie , Cellules A549 , Carcinome pulmonaire non à petites cellules/génétique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Cétuximab/pharmacologie , Récepteurs ErbB/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des radiations , Humains , Tumeurs du poumon/génétique , Mâle , Thérapie moléculaire ciblée , Nanoparticules , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protéines proto-oncogènes/antagonistes et inhibiteurs , Petit ARN interférent/pharmacologie , Radiosensibilisants/pharmacologie , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe ,
2.
Bioengineering (Basel) ; 6(1)2019 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-30875927

RÉSUMÉ

Multiparametric and high-content protein analysis of single cells or tissues cannot be accomplished with the currently available flow cytometry or imaging techniques utilizing fluorophore-labelled antibodies, because the number of spectrally resolvable fluorochromes is limited. In contrast, mass cytometry can resolve more signals by exploiting lanthanide-tagged antibodies; however, only about 100 metal reporters can be attached to an antibody molecule. This makes the sensitivity of lanthanide-tagged antibodies substantially lower than fluorescent reporters. A new probe that can carry more lanthanide molecules per antibody is a desirable way to enhance the sensitivity needed for the detection of protein with low cellular abundance. Herein, we report on the development of new probes utilizing mesoporous silica nanoparticles (MSNPs) with hydroxyl, amine, or phosphonate functional groups. The phosphonated MSNPs proved to be best at loading lanthanides for up to 1.4 × 106 molecules per particle, and could be loaded with various lanthanide elements (Ce, Pr, Nd, Sm, Eu, Gd, Tb, Dy, Ho, Er, Yb, and Lu) at relatively similar molar extents. The modified MSNPs can also load a fluorescent dye, allowing bimodal mass and fluorescence-based detection. We achieved specificity of antibody-conjugated nanoparticles (at 1.4 × 10³ antibodies per nanoparticle) for targeting proteins on the cell surface. The new materials can potentially be used as mass cytometry probes and provide a method for simultaneous monitoring of a large host of factors comprising the tumor microenvironment (e.g., extracellular matrix, cancer cells, and immune cells). These novel probes may also benefit personalized medicine by allowing for high-throughput analysis of multiple proteins in the same specimen.

3.
Int J Nanomedicine ; 13: 4015-4027, 2018.
Article de Anglais | MEDLINE | ID: mdl-30022824

RÉSUMÉ

INTRODUCTION: Long-term stability of therapeutic candidates is necessary toward their clinical applications. For most nanoparticle systems formulated in aqueous solutions, lyophilization or freeze-drying is a common method to ensure long-term stability. While lyophilization of lipid, polymeric, or inorganic nanoparticles have been studied, little has been reported on lyophilization and stability of hybrid nanoparticle systems, consisting of polymers, inorganic particles, and antibody. Lyophilization of complex nanoparticle systems can be challenging with respect to preserving physicochemical properties and the biological activities of the materials. We recently reported an effective small-interfering RNA (siRNA) nanoparticle carrier consisting of 50-nm mesoporous silica nanoparticles decorated with a copolymer of polyethylenimine and polyethyleneglycol, and antibody. MATERIALS AND METHODS: Toward future personalized medicine, the nanoparticle carriers were lyophilized alone and loaded with siRNA upon reconstitution by a few minutes of simple mixing in phosphate-buffered saline. Herein, we optimize the lyophilization of the nanoparticles in terms of buffers, lyoprotectants, reconstitution, and time and temperature of freezing and drying steps, and monitor the physical and chemical properties (reconstitution, hydrodynamic size, charge, and siRNA loading) and biological activities (gene silencing, cancer cell killing) of the materials after storing at various temperatures and times. RESULTS: The material was best formulated in Tris-HCl buffer with 5% w/w trehalose. Freezing step was performed at -55°C for 3 h, followed by a primary drying step at -40°C (100 µBar) for 24 h and a secondary drying step at 20°C (20 µBar) for 12 h. The lyophilized material can be stored stably for 2 months at 4°C and at least 6 months at -20°C. CONCLUSION: We successfully developed the lyophilization process that should be applicable to other similar nanoparticle systems consisting of inorganic nanoparticle cores modified with cationic polymers, PEG, and antibodies.


Sujet(s)
Anticorps/composition chimique , Lyophilisation/méthodes , Nanoparticules/composition chimique , Polyéthylène glycols/composition chimique , Polyéthylèneimine/composition chimique , Petit ARN interférent/administration et posologie , Silice/composition chimique , Substances tampon , Cations , Lignée cellulaire tumorale , Stabilité de médicament , Congélation , Humains , Hydrodynamique , Taille de particule , Porosité , Température
4.
PLoS One ; 13(6): e0198141, 2018.
Article de Anglais | MEDLINE | ID: mdl-29879129

RÉSUMÉ

Intrinsic and acquired resistance to current HER2 targeted therapies remains a challenge in clinics. We have developed a therapeutic HER2 siRNA delivered using mesoporous silica nanoparticles modified with polymers and conjugated with HER2 targeting antibodies. Our previous studies have shown that our HER2 siRNA nanoparticles could overcome intrinsic and acquired resistance to trastuzumab and lapatinib in HER2-positive breast cancers. In this study, we investigated the effect of long-term (7 months) treatment using our therapeutic HER2 siRNA. Even after the removal of HER2 siRNA, the long-term treated cells grew much slower (67% increase in doubling time) than cells that have not received any treatment. The treated cells did not undergo epithelial-mesenchymal transition or showed enrichment of tumor initiating cells. Unlike trastuzumab and lapatinib, which induced resistance in BT474 cells after 6 months of treatment, HER2 siRNA did not induce resistance to HER2 siRNA, trastuzumab, or lapatinib. HER2 ablation with HER2 siRNA prevented reactivation of HER2 signaling that was observed in cells resistant to lapatinib. Altogether, our results indicate that a HER2 siRNA based therapeutic provides a more durable inhibition of HER2 signaling in vitro and can potentially be more effective than the existing therapeutic monoclonal antibodies and small molecule inhibitors.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Petit ARN interférent/usage thérapeutique , Récepteur ErbB-2/génétique , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Femelle , Humains , Lapatinib/usage thérapeutique , Thérapie moléculaire ciblée/effets indésirables , Nanoparticules/usage thérapeutique , Petit ARN interférent/génétique , Petit ARN interférent/pharmacologie , Récepteur ErbB-2/antagonistes et inhibiteurs , Trastuzumab/usage thérapeutique
6.
Mol Cancer Ther ; 16(4): 763-772, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28138033

RÉSUMÉ

Metastatic breast cancer is developed in about 20% to 30% of newly diagnosed patients with early-stage breast cancer despite treatments. Herein, we report a novel nanoparticle platform with intrinsic antimetastatic properties for the targeted delivery of Polo-like kinase 1 siRNA (siPLK1). We first evaluated it in a triple-negative breast cancer (TNBC) model, which shows high metastatic potential. PLK1 was identified as the top therapeutic target for TNBC cells and tumor-initiating cells in a kinome-wide screen. The platform consists of a 50-nm mesoporous silica nanoparticle (MSNP) core coated layer-by-layer with bioreducible cross-linked PEI and PEG polymers, conjugated with an antibody for selective uptake into cancer cells. siRNA is loaded last and fully protected under the PEG layer from blood enzymatic degradation. The material has net neutral charge and low nonspecific cytotoxicity. We have also shown for the first time that the MSNP itself inhibited cancer migration and invasion in TNBC cells owing to its ROS- and NOX4-modulating properties. In vivo, siPLK1 nanoconstructs (six doses of 0.5 mg/kg) knocked down about 80% of human PLK1 mRNA expression in metastatic breast cancer cells residing in mouse lungs and reduced tumor incidence and burden in lungs and other organs of an experimental metastasis mouse model. Long-term treatment significantly delayed the onset of death in mice and improved the overall survival. The platform capable of simultaneously inhibiting the proliferative and metastatic hallmarks of cancer progression is unique and has great therapeutic potential to also target other metastatic cancers beyond TNBC. Mol Cancer Ther; 16(4); 763-72. ©2017 AACR.


Sujet(s)
Antioxydants/administration et posologie , Protéines du cycle cellulaire/génétique , Nanoparticules/administration et posologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes/génétique , Petit ARN interférent/administration et posologie , Tumeurs du sein triple-négatives/thérapie , Animaux , Antioxydants/pharmacologie , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Souris , Métastase tumorale , Cellules souches tumorales/métabolisme , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protéines proto-oncogènes/antagonistes et inhibiteurs , Tumeurs du sein triple-négatives/génétique , Tests d'activité antitumorale sur modèle de xénogreffe ,
7.
Breast Cancer Res ; 18(1): 70, 2016 07 01.
Article de Anglais | MEDLINE | ID: mdl-27368372

RÉSUMÉ

BACKGROUND: High mitotic activity is associated with the genesis and progression of many cancers. Small molecule inhibitors of mitotic apparatus proteins are now being developed and evaluated clinically as anticancer agents. With clinical trials of several of these experimental compounds underway, it is important to understand the molecular mechanisms that determine high mitotic activity, identify tumor subtypes that carry molecular aberrations that confer high mitotic activity, and to develop molecular markers that distinguish which tumors will be most responsive to mitotic apparatus inhibitors. METHODS: We identified a coordinately regulated mitotic apparatus network by analyzing gene expression profiles for 53 malignant and non-malignant human breast cancer cell lines and two separate primary breast tumor datasets. We defined the mitotic network activity index (MNAI) as the sum of the transcriptional levels of the 54 coordinately regulated mitotic apparatus genes. The effect of those genes on cell growth was evaluated by small interfering RNA (siRNA). RESULTS: High MNAI was enriched in basal-like breast tumors and was associated with reduced survival duration and preferential sensitivity to inhibitors of the mitotic apparatus proteins, polo-like kinase, centromere associated protein E and aurora kinase designated GSK462364, GSK923295 and GSK1070916, respectively. Co-amplification of regions of chromosomes 8q24, 10p15-p12, 12p13, and 17q24-q25 was associated with the transcriptional upregulation of this network of 54 mitotic apparatus genes, and we identify transcription factors that localize to these regions and putatively regulate mitotic activity. Knockdown of the mitotic network by siRNA identified 22 genes that might be considered as additional therapeutic targets for this clinically relevant patient subgroup. CONCLUSIONS: We define a molecular signature which may guide therapeutic approaches for tumors with high mitotic network activity.


Sujet(s)
Tumeurs du sein/génétique , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique/génétique , Génome humain/génétique , Mitose/effets des médicaments et des substances chimiques , Aurora kinases/antagonistes et inhibiteurs , Aurora kinases/génétique , Aurora kinases/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Protéines chromosomiques nonhistones/antagonistes et inhibiteurs , Protéines chromosomiques nonhistones/génétique , Protéines chromosomiques nonhistones/métabolisme , Femelle , Amplification de gène , Analyse de profil d'expression de gènes/méthodes , Réseaux de régulation génique/effets des médicaments et des substances chimiques , Humains , Estimation de Kaplan-Meier , Mitose/génétique , Pronostic , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Interférence par ARN , Bibliothèques de petites molécules/pharmacologie , Résultat thérapeutique ,
8.
Cancer Treat Rev ; 45: 19-29, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26930249

RÉSUMÉ

This Review discusses the various types of non-coding oligonucleotides, which have garnered extensive interest as new alternatives for targeted cancer therapies over small molecule inhibitors and monoclonal antibodies. These oligonucleotides can target any hallmark of cancer, no longer limited to so-called "druggable" targets. Thus, any identified gene that plays a key role in cancer progression or drug resistance can be exploited with oligonucleotides. Among them, small-interfering RNAs (siRNAs) are frequently utilized for gene silencing due to the robust and well established mechanism of RNA interference. Despite promising advantages, clinical translation of siRNAs is hindered by the lack of effective delivery platforms. This Review provides general criteria and consideration of nanoparticle development for systemic siRNA delivery. Different classes of nanoparticle candidates for siRNA delivery are discussed, and the progress in clinical trials for systemic cancer treatment is reviewed. Lastly, this Review presents HER2 (human epidermal growth factor receptor type 2)-positive breast cancer as one example that could benefit significantly from siRNA technology. How siRNA-based therapeutics can overcome cancer resistance to such therapies is discussed.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Oligonucléotides/pharmacologie , Petit ARN interférent/pharmacologie , Récepteur ErbB-2/génétique , Administration par voie intraveineuse/méthodes , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Systèmes de délivrance de médicaments/méthodes , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Humains , Mâle , Nanoparticules , Thérapie par l'interférence par ARN/méthodes
9.
Oncotarget ; 7(12): 14727-41, 2016 Mar 22.
Article de Anglais | MEDLINE | ID: mdl-26894975

RÉSUMÉ

HER2 is overexpressed in about 20% of breast cancers and contributes to poor prognosis. Unfortunately, a large fraction of patients have primary or acquired resistance to the HER2-targeted therapy trastuzumab, thus a multi-drug combination is utilized in the clinic, putting significant burden on patients. We systematically identified an optimal HER2 siRNA from 76 potential sequences and demonstrated its utility in overcoming intrinsic and acquired resistance to trastuzumab and lapatinib in 18 HER2-positive cancer cell lines. We provided evidence that the drug-resistant cancer maintains dependence on HER2 for survival. Importantly, cell lines did not readily develop resistance following extended treatment with HER2 siRNA. Using our recently developed nanoparticle platform, systemic delivery of HER2 siRNA to trastuzumab-resistant tumors resulted in significant growth inhibition. Moreover, the optimal HER2 siRNA could also silence an exon 16 skipped HER2 splice variant reported to be highly oncogenic and linked to trastuzumab resistance.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Petit ARN interférent/génétique , Récepteur ErbB-2/antagonistes et inhibiteurs , Animaux , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Lapatinib , Souris , Souris nude , Phosphorylation , Quinazolines/pharmacologie , Récepteur ErbB-2/génétique , Transduction du signal , Trastuzumab/pharmacologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Biomaterials ; 66: 41-52, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26196532

RÉSUMÉ

Fibrotic diseases such as scleroderma have been linked to increased oxidative stress and upregulation of pro-fibrotic genes. Recent work suggests a role of NADPH oxidase 4 (NOX4) and heat shock protein 47 (HSP47) in inducing excessive collagen synthesis, leading to fibrotic diseases. Herein, we elucidate the relationship between NOX4 and HSP47 in fibrogenesis and propose to modulate them altogether as a new strategy to treat fibrosis. We developed a nanoparticle platform consisting of polyethylenimine (PEI) and polyethylene glycol (PEG) coating on a 50-nm mesoporous silica nanoparticle (MSNP) core. The nanoparticles effectively delivered small interfering RNA (siRNA) targeting HSP47 (siHSP47) in an in vitro model of fibrosis based on TGF-ß stimulated fibroblasts. The MSNP core also imparted an antioxidant property by scavenging reactive oxygen species (ROS) and subsequently reducing NOX4 levels in the in vitro fibrogenesis model. The nanoparticle was far superior to n-acetyl cysteine (NAC) at modulating pro-fibrotic markers. In vivo evaluation was performed in a bleomycin-induced scleroderma mouse model, which shares many similarities to human scleroderma disease. Intradermal administration of siHSP47-nanoparticles effectively reduced HSP47 protein expression in skin to normal level. In addition, the antioxidant MSNP also played a prominent role in reducing the pro-fibrotic markers, NOX4, alpha smooth muscle actin (α-SMA), and collagen type I (COL I), as well as skin thickness of the mice.


Sujet(s)
Protéines de choc thermique HSP47/génétique , NADPH oxidase/génétique , Nanocapsules/composition chimique , Petit ARN interférent/génétique , Sclérodermie diffuse/génétique , Sclérodermie diffuse/thérapie , Administration par voie cutanée , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Extinction de l'expression des gènes , Thérapie génétique/méthodes , Souris , Souris de lignée C3H , NADPH Oxidase 4 , Nanocapsules/administration et posologie , Nanocapsules/ultrastructure , Nanopores/ultrastructure , Taille de particule , Porosité , Petit ARN interférent/administration et posologie , Silice/composition chimique , Résultat thérapeutique
11.
Adv Funct Mater ; 25(18): 2646-2659, 2015 May 13.
Article de Anglais | MEDLINE | ID: mdl-26097445

RÉSUMÉ

In vivo delivery of siRNAs designed to inhibit genes important in cancer and other diseases continues to be an important biomedical goal. We now describe a new nanoparticle construct that has been engineered for efficient delivery of siRNA to tumors. The construct is comprised of a 47-nm mesoporous silica nanoparticle (MSNP) core coated with a cross-linked PEI-PEG copolymer, carrying siRNA against the HER2 oncogene, and coupled to the anti-HER2 monoclonal antibody (trastuzumab). The construct has been engineered to increase siRNA blood half-life, enhance tumor-specific cellular uptake, and maximize siRNA knockdown efficacy. The optimized anti-HER2-nanoparticles produced apoptotic death in HER2 positive (HER2+) breast cancer cells grown in vitro, but not in HER2 negative (HER2-) cells. One dose of the siHER2-nanoparticles reduced HER2 protein levels by 60% in trastuzumab-resistant HCC1954 xenografts. Multiple doses administered intravenously over 3 weeks significantly inhibited tumor growth (p < 0.004). The siHER2-nanoparticles have an excellent safety profile in terms of blood compatibility and low cytokine induction, when exposed to human peripheral blood mononuclear cells. The construct can be produced with high batch-to-batch reproducibility and the production methods are suitable for large-scale production. These results suggest that this siHER2-nanoparticle is ready for clinical evaluation.

12.
Theranostics ; 4(9): 872-92, 2014.
Article de Anglais | MEDLINE | ID: mdl-25057313

RÉSUMÉ

RNA interference (RNAi) is an endogenous post-transcriptional gene regulatory mechanism, where non-coding, double-stranded RNA molecules interfere with the expression of certain genes in order to silence it. Since its discovery, this phenomenon has evolved as powerful technology to diagnose and treat diseases at cellular and molecular levels. With a lot of attention, short interfering RNA (siRNA) therapeutics has brought a great hope for treatment of various undruggable diseases, including genetic diseases, cancer, and resistant viral infections. However, the challenge of their systemic delivery and on how they are integrated to exhibit the desired properties and functions remains a key bottleneck for realizing its full potential. Nanoparticles are currently well known to exhibit a number of unique properties that could be strategically tailored into new advanced siRNA delivery systems. This review summarizes the various nanoparticulate systems developed so far in the literature for systemic delivery of siRNA, which include silica and silicon-based nanoparticles, metal and metal oxides nanoparticles, carbon nanotubes, graphene, dendrimers, polymers, cyclodextrins, lipids, hydrogels, and semiconductor nanocrystals. Challenges and barriers to the delivery of siRNA and the role of different nanoparticles to surmount these challenges are also included in the review.


Sujet(s)
Nanoparticules , Tumeurs/traitement médicamenteux , Petit ARN interférent/usage thérapeutique , Maladies virales/traitement médicamenteux , Animaux , Systèmes de délivrance de médicaments , Humains
13.
Cancer Discov ; 2(8): 685-93, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22628411

RÉSUMÉ

UNLABELLED: KRAS mutation is a hallmark of pancreatic ductal adenocarcinoma (PDA) but remains an intractable pharmacologic target. Consequently, defining RAS effector pathway(s) required for PDA initiation and maintenance is critical to improve treatment of this disease. Here, we show that expression of BRAF(V600E), but not PIK3CA(H1047R), in the mouse pancreas leads to pancreatic intraepithelial neoplasia (PanIN) lesions. Moreover, concomitant expression of BRAF(V600E) and TP53(R270H) result in lethal PDA. We tested pharmacologic inhibitors of RAS effectors against multiple human PDA cell lines. Mitogen-activated protein (MAP)/extracellular signal-regulated (ERK) kinase (MEK) inhibition was highly effective both in vivo and in vitro and was synergistic with AKT inhibition in most cell lines tested. We show that RAF→MEK→ERK signaling is central to the initiation and maintenance of PDA and to rational combination strategies in this disease. These results emphasize the value of leveraging multiple complementary experimental systems to prioritize pathways for effective intervention strategies in PDA. SIGNIFICANCE: PDA is diffi cult to treat, in large part, due to recurrent mutations in the KRAS gene. Here, we defi ne rational treatment approaches for the disease achievable today with existing drug combinations by thorough genetic and pharmacologic dissection of the major KRAS effector pathways, RAF→MEK→ERK and phosphoinositide 3'-kinase (PI3'K)→AKT.


Sujet(s)
Carcinome du canal pancréatique/enzymologie , Transformation cellulaire néoplasique/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Système de signalisation des MAP kinases , Mitogen-Activated Protein Kinase Kinases/métabolisme , Tumeurs du pancréas/enzymologie , Protéines proto-oncogènes B-raf/métabolisme , Animaux , Carcinome du canal pancréatique/génétique , Lignée cellulaire tumorale , Transformation cellulaire néoplasique/génétique , Extracellular Signal-Regulated MAP Kinases/génétique , Gènes ras , Humains , Immunotransfert , Immunohistochimie , Souris , Mitogen-Activated Protein Kinase Kinases/génétique , Tumeurs du pancréas/génétique , Protéines proto-oncogènes B-raf/génétique
14.
Nat Med ; 17(4): 500-3, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-21460848

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDA) is a lethal disease. Overall survival is typically 6 months from diagnosis. Numerous phase 3 trials of agents effective in other malignancies have failed to benefit unselected PDA populations, although patients do occasionally respond. Studies in other solid tumors have shown that heterogeneity in response is determined, in part, by molecular differences between tumors. Furthermore, treatment outcomes are improved by targeting drugs to tumor subtypes in which they are selectively effective, with breast and lung cancers providing recent examples. Identification of PDA molecular subtypes has been frustrated by a paucity of tumor specimens available for study. We have overcome this problem by combined analysis of transcriptional profiles of primary PDA samples from several studies, along with human and mouse PDA cell lines. We define three PDA subtypes: classical, quasimesenchymal and exocrine-like, and we present evidence for clinical outcome and therapeutic response differences between them. We further define gene signatures for these subtypes that may have utility in stratifying patients for treatment and present preclinical model systems that may be used to identify new subtype specific therapies.


Sujet(s)
Carcinome du canal pancréatique/classification , Carcinome du canal pancréatique/génétique , Tumeurs du pancréas/classification , Tumeurs du pancréas/génétique , Animaux , Antinéoplasiques/pharmacologie , Carcinome du canal pancréatique/traitement médicamenteux , Lignée cellulaire tumorale , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Chlorhydrate d'erlotinib , Femelle , Facteur de transcription GATA-6/génétique , Analyse de profil d'expression de gènes , Humains , Mâle , Souris , Tumeurs du pancréas/traitement médicamenteux , Pharmacogénétique , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes p21(ras) , Quinazolines/pharmacologie , Protéines G ras/génétique ,
15.
Breast Cancer Res ; 12(2): R18, 2010.
Article de Anglais | MEDLINE | ID: mdl-20211017

RÉSUMÉ

INTRODUCTION: HJURP (Holliday Junction Recognition Protein) is a newly discovered gene reported to function at centromeres and to interact with CENPA. However its role in tumor development remains largely unknown. The goal of this study was to investigate the clinical significance of HJURP in breast cancer and its correlation with radiotherapeutic outcome. METHODS: We measured HJURP expression level in human breast cancer cell lines and primary breast cancers by Western blot and/or by Affymetrix Microarray; and determined its associations with clinical variables using standard statistical methods. Validation was performed with the use of published microarray data. We assessed cell growth and apoptosis of breast cancer cells after radiation using high-content image analysis. RESULTS: HJURP was expressed at higher level in breast cancer than in normal breast tissue. HJURP mRNA levels were significantly associated with estrogen receptor (ER), progesterone receptor (PR), Scarff-Bloom-Richardson (SBR) grade, age and Ki67 proliferation indices, but not with pathologic stage, ERBB2, tumor size, or lymph node status. Higher HJURP mRNA levels significantly decreased disease-free and overall survival. HJURP mRNA levels predicted the prognosis better than Ki67 proliferation indices. In a multivariate Cox proportional-hazard regression, including clinical variables as covariates, HJURP mRNA levels remained an independent prognostic factor for disease-free and overall survival. In addition HJURP mRNA levels were an independent prognostic factor over molecular subtypes (normal like, luminal, Erbb2 and basal). Poor clinical outcomes among patients with high HJURP expression were validated in five additional breast cancer cohorts. Furthermore, the patients with high HJURP levels were much more sensitive to radiotherapy. In vitro studies in breast cancer cell lines showed that cells with high HJURP levels were more sensitive to radiation treatment and had a higher rate of apoptosis than those with low levels. Knock down of HJURP in human breast cancer cells using shRNA reduced the sensitivity to radiation treatment. HJURP mRNA levels were significantly correlated with CENPA mRNA levels. CONCLUSIONS: HJURP mRNA level is a prognostic factor for disease-free and overall survival in patients with breast cancer and is a predictive biomarker for sensitivity to radiotherapy.


Sujet(s)
Tumeurs du sein/radiothérapie , Protéines de liaison à l'ADN/génétique , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/effets des radiations , Marqueurs biologiques tumoraux/analyse , Technique de Western , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/métabolisme , Survie sans rechute , Femelle , Humains , Séquençage par oligonucléotides en batterie , Valeur prédictive des tests , Pronostic , Interférence par ARN , ARN messager/génétique , ARN messager/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...