Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
1.
Nat Commun ; 15(1): 5763, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982051

RÉSUMÉ

While high circulating tumor DNA (ctDNA) levels are associated with poor survival for multiple cancers, variant-specific differences in the association of ctDNA levels and survival have not been examined. Here we investigate KRAS ctDNA (ctKRAS) variant-specific associations with overall and progression-free survival (OS/PFS) in first-line metastatic pancreatic ductal adenocarcinoma (mPDAC) for patients receiving chemoimmunotherapy ("PRINCE", NCT03214250), and an independent cohort receiving standard of care (SOC) chemotherapy. For PRINCE, higher baseline plasma levels are associated with worse OS for ctKRAS G12D (log-rank p = 0.0010) but not G12V (p = 0.7101), even with adjustment for clinical covariates. Early, on-therapy clearance of G12D (p = 0.0002), but not G12V (p = 0.4058), strongly associates with OS for PRINCE. Similar results are obtained for the SOC cohort, and for PFS in both cohorts. These results suggest ctKRAS G12D but not G12V as a promising prognostic biomarker for mPDAC and that G12D clearance could also serve as an early biomarker of response.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome du canal pancréatique , ADN tumoral circulant , Tumeurs du pancréas , Protéines proto-oncogènes p21(ras) , Humains , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/mortalité , Carcinome du canal pancréatique/sang , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/traitement médicamenteux , Protéines proto-oncogènes p21(ras)/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/sang , Tumeurs du pancréas/mortalité , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/traitement médicamenteux , Femelle , Mâle , ADN tumoral circulant/sang , ADN tumoral circulant/génétique , Adulte d'âge moyen , Sujet âgé , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/génétique , Pronostic , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Mutation , Survie sans progression , Métastase tumorale
2.
Front Oncol ; 14: 1343004, 2024.
Article de Anglais | MEDLINE | ID: mdl-38371625

RÉSUMÉ

MCL1 is a member of the BCL2 family of apoptosis regulators, which play a critical role in promoting cancer survival and drug resistance. We previously described PRT1419, a potent, MCL1 inhibitor with anti-tumor efficacy in various solid and hematologic malignancies. To identify novel biomarkers that predict sensitivity to MCL1 inhibition, we conducted a gene essentiality analysis using gene dependency data generated from CRISPR/Cas9 cell viability screens. We observed that clear cell renal cancer (ccRCC) cell lines with damaging PBRM1 mutations displayed a strong dependency on MCL1. PBRM1 (BAF180), is a chromatin-targeting subunit of mammalian pBAF complexes. PBRM1 is frequently altered in various cancers particularly ccRCC with ~40% of tumors harboring damaging PBRM1 alterations. We observed potent inhibition of tumor growth and induction of apoptosis by PRT1419 in various preclinical models of PBRM1-mutant ccRCC but not PBRM1-WT. Depletion of PBRM1 in PBRM1-WT ccRCC cell lines induced sensitivity to PRT1419. Mechanistically, PBRM1 depletion coincided with increased expression of pro-apoptotic factors, priming cells for caspase-mediated apoptosis following MCL1 inhibition. Increased MCL1 activity has been described as a resistance mechanism to Sunitinib and Everolimus, two approved agents for ccRCC. PRT1419 synergized with both agents to potently inhibit tumor growth in PBRM1-loss ccRCC. PRT2527, a potent CDK9 inhibitor which depletes MCL1, was similarly efficacious in monotherapy and in combination with Sunitinib in PBRM1-loss cells. Taken together, these findings suggest PBRM1 loss is associated with MCL1i sensitivity in ccRCC and provide rationale for the evaluation of PRT1419 and PRT2527 for the treatment for PBRM1-deficient ccRCC.

3.
Cell Rep ; 43(1): 113608, 2024 01 23.
Article de Anglais | MEDLINE | ID: mdl-38117649

RÉSUMÉ

The role of hematopoietic Hedgehog signaling in myeloproliferative neoplasms (MPNs) remains incompletely understood despite data suggesting that Hedgehog (Hh) pathway inhibitors have therapeutic activity in patients. We aim to systematically interrogate the role of canonical vs. non-canonical Hh signaling in MPNs. We show that Gli1 protein levels in patient peripheral blood mononuclear cells (PBMCs) mark fibrotic progression and that, in murine MPN models, absence of hematopoietic Gli1, but not Gli2 or Smo, significantly reduces MPN phenotype and fibrosis, indicating that GLI1 in the MPN clone can be activated in a non-canonical fashion. Additionally, we establish that hematopoietic Gli1 has a significant effect on stromal cells, mediated through a druggable MIF-CD74 axis. These data highlight the complex interplay between alterations in the MPN clone and activation of stromal cells and indicate that Gli1 represents a promising therapeutic target in MPNs, particularly that Hh signaling is dispensable for normal hematopoiesis.


Sujet(s)
Antinéoplasiques , Syndromes myéloprolifératifs , Tumeurs , Humains , Souris , Animaux , Protéines Hedgehog/métabolisme , Protéine à doigt de zinc GLI1/métabolisme , Agranulocytes/métabolisme , Hématopoïèse
4.
Cancer Res Commun ; 3(11): 2233-2243, 2023 11 06.
Article de Anglais | MEDLINE | ID: mdl-37861290

RÉSUMÉ

Expression of protein arginine methyltransferase 5 (PRMT5) is highly positively correlated to DNA damage repair (DDR) and DNA replication pathway genes in many types of cancer cells, including ovarian and breast cancer. In the current study, we investigated whether pharmacologic inhibition of PRMT5 downregulates DDR/DNA replication pathway genes and sensitizes cancer cells to chemotherapy and PARP inhibition. Potent and selective PRMT5 inhibitors significantly downregulate expression of multiple DDR and DNA replication genes in cancer cells. Mechanistically, PRMT5 inhibition reduces the presence of PRMT5 and H4R3me2s on promoter regions of DDR genes such as BRCA1/2, RAD51, and ATM. PRMT5 inhibition also promotes global alternative splicing changes. Our data suggest that PRMT5 inhibition regulates expression of FANCA, PNKP, and ATM by promoting exon skipping and intron retention. Combining C220 or PRT543 with olaparib or chemotherapeutic agents such as cisplatin demonstrates a potent synergistic interaction in breast and ovarian cancer cells in vitro. Moreover, combination of PRT543 with olaparib effectively inhibits the growth of patient-derived breast and ovarian cancer xenografts. Furthermore, PRT543 treatment significantly inhibits growth of olaparib-resistant tumors in vivo. These studies reveal a novel mechanism of PRMT5 inhibition and suggest beneficial combinatorial effects with other therapies, particularly in patients with tumors that are resistant to therapies dependent on DNA damage as their mechanism of action. SIGNIFICANCE: Patients with advanced cancers frequently develop resistance to chemotherapy or PARP inhibitors mainly due to circumvention and/or restoration of the inactivated DDR pathway genes. We demonstrate that inhibition of PRMT5 significantly downregulates a broad range of the DDR and DNA replication pathway genes. PRMT5 inhibitors combined with chemotherapy or PARP inhibitors demonstrate synergistic suppression of cancer cell proliferation and growth in breast and ovarian tumor models, including PARP inhibitor-resistant tumors.


Sujet(s)
Tumeurs du sein , Tumeurs de l'ovaire , Humains , Femelle , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Antienzymes , Altération de l'ADN , Tumeurs du sein/traitement médicamenteux , Tumeurs de l'ovaire/traitement médicamenteux , Protein-arginine N-methyltransferases/génétique , Phosphotransferases (Alcohol Group Acceptor)/génétique , Enzymes de réparation de l'ADN/génétique
5.
Blood Adv ; 7(20): 6211-6224, 2023 Oct 24.
Article de Anglais | MEDLINE | ID: mdl-37327122

RÉSUMÉ

Mantle cell lymphoma (MCL) is an incurable B-cell malignancy that comprises up to 6% of non-Hodgkin lymphomas diagnosed annually and is associated with a poor prognosis. The average overall survival of patients with MCL is 5 years, and for most patients who progress on targeted agents, survival remains at a dismal 3 to 8 months. There is a major unmet need to identify new therapeutic approaches that are well tolerated to improve treatment outcomes and quality of life. The protein arginine methyltransferase 5 (PRMT5) enzyme is overexpressed in MCL and promotes growth and survival. Inhibition of PRMT5 drives antitumor activity in MCL cell lines and preclinical murine models. PRMT5 inhibition reduced the activity of prosurvival AKT signaling, which led to the nuclear translocation of FOXO1 and modulation of its transcriptional activity. Chromatin immunoprecipitation and sequencing identified multiple proapoptotic BCL-2 family members as FOXO1-bound genomic loci. We identified BAX as a direct transcriptional target of FOXO1 and demonstrated its critical role in the synergy observed between the selective PRMT5 inhibitor, PRT382, and the BCL-2 inhibitor, venetoclax. Single-agent and combination treatments were performed in 9 MCL lines. Loewe synergy scores showed significant levels of synergy in most MCL lines tested. Preclinical, in vivo evaluation of this strategy in multiple MCL models showed therapeutic synergy with combination venetoclax/PRT382 treatment with an increased survival advantage in 2 patient-derived xenograft models (P ≤ .0001, P ≤ .0001). Our results provide mechanistic rationale for the combination of PRMT5 inhibition and venetoclax to treat patients with MCL.


Sujet(s)
Antinéoplasiques , Composés hétérocycliques bicycliques , Lymphome à cellules du manteau , Sulfonamides , Animaux , Humains , Souris , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Lymphome à cellules du manteau/traitement médicamenteux , Lymphome à cellules du manteau/génétique , Lymphome à cellules du manteau/métabolisme , Protein-arginine N-methyltransferases/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Qualité de vie
6.
Cancer Discov ; 10(11): 1742-1757, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32669286

RÉSUMÉ

We investigated the role of PRMT5 in myeloproliferative neoplasm (MPN) pathogenesis and aimed to elucidate key PRMT5 targets contributing to MPN maintenance. PRMT5 is overexpressed in primary MPN cells, and PRMT5 inhibition potently reduced MPN cell proliferation ex vivo. PRMT5 inhibition was efficacious at reversing elevated hematocrit, leukocytosis, and splenomegaly in a model of JAK2V617F+ polycythemia vera and leukocyte and platelet counts, hepatosplenomegaly, and fibrosis in the MPLW515L model of myelofibrosis. Dual targeting of JAK and PRMT5 was superior to JAK or PRMT5 inhibitor monotherapy, further decreasing elevated counts and extramedullary hematopoiesis in vivo. PRMT5 inhibition reduced expression of E2F targets and altered the methylation status of E2F1 leading to attenuated DNA damage repair, cell-cycle arrest, and increased apoptosis. Our data link PRMT5 to E2F1 regulatory function and MPN cell survival and provide a strong mechanistic rationale for clinical trials of PRMT5 inhibitors in MPN. SIGNIFICANCE: Expression of PRMT5 and E2F targets is increased in JAK2V617F+ MPN. Pharmacologic inhibition of PRMT5 alters the methylation status of E2F1 and shows efficacy in JAK2V617F/MPLW515L MPN models and primary samples. PRMT5 represents a potential novel therapeutic target for MPN, which is now being clinically evaluated.This article is highlighted in the In This Issue feature, p. 1611.


Sujet(s)
Facteur de transcription E2F1/métabolisme , Réseaux de régulation génique/génétique , Kinase Janus-2/métabolisme , Protein-arginine N-methyltransferases/antagonistes et inhibiteurs , Humains , Méthylation , Mutation , Protein-arginine N-methyltransferases/métabolisme
7.
Clin Cancer Res ; 26(13): 3248-3258, 2020 07 01.
Article de Anglais | MEDLINE | ID: mdl-32299821

RÉSUMÉ

PURPOSE: To determine whether a multianalyte liquid biopsy can improve the detection and staging of pancreatic ductal adenocarcinoma (PDAC). EXPERIMENTAL DESIGN: We analyzed plasma from 204 subjects (71 healthy, 44 non-PDAC pancreatic disease, and 89 PDAC) for the following biomarkers: tumor-associated extracellular vesicle miRNA and mRNA isolated on a nanomagnetic platform that we developed and measured by next-generation sequencing or qPCR, circulating cell-free DNA (ccfDNA) concentration measured by qPCR, ccfDNA KRAS G12D/V/R mutations detected by droplet digital PCR, and CA19-9 measured by electrochemiluminescence immunoassay. We applied machine learning to training sets and subsequently evaluated model performance in independent, user-blinded test sets. RESULTS: To identify patients with PDAC versus those without, we generated a classification model using a training set of 47 subjects (20 PDAC and 27 noncancer). When applied to a blinded test set (N = 136), the model achieved an AUC of 0.95 and accuracy of 92%, superior to the best individual biomarker, CA19-9 (89%). We next used a cohort of 20 patients with PDAC to train our model for disease staging and applied it to a blinded test set of 25 patients clinically staged by imaging as metastasis-free, including 9 subsequently determined to have had occult metastasis. Our workflow achieved significantly higher accuracy for disease staging (84%) than imaging alone (accuracy = 64%; P < 0.05). CONCLUSIONS: Algorithmically combining blood-based biomarkers may improve PDAC diagnostic accuracy and preoperative identification of nonmetastatic patients best suited for surgery, although larger validation studies are necessary.


Sujet(s)
Adénocarcinome/diagnostic , Adénocarcinome/métabolisme , Marqueurs biologiques tumoraux , Vésicules extracellulaires/métabolisme , Tumeurs du pancréas/diagnostic , Tumeurs du pancréas/métabolisme , Adénocarcinome/étiologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigène CA 19-9 , Acides nucléiques acellulaires , Biologie informatique/méthodes , Femelle , Humains , Biopsie liquide/méthodes , Apprentissage machine , Mâle , microARN , Adulte d'âge moyen , Mutation , Stadification tumorale , Tumeurs du pancréas/étiologie , Protéines proto-oncogènes p21(ras)/génétique , ARN messager , Courbe ROC
9.
Nucleic Acids Res ; 48(4): e19, 2020 02 28.
Article de Anglais | MEDLINE | ID: mdl-31828328

RÉSUMÉ

Detection of disease-associated, cell-free nucleic acids in body fluids enables early diagnostics, genotyping and personalized therapy, but is challenged by the low concentrations of clinically significant nucleic acids and their sequence homology with abundant wild-type nucleic acids. We describe a novel approach, dubbed NAVIGATER, for increasing the fractions of Nucleic Acids of clinical interest Via DNA-Guided Argonaute from Thermus thermophilus (TtAgo). TtAgo cleaves specifically guide-complementary DNA and RNA with single nucleotide precision, greatly increasing the fractions of rare alleles and, enhancing the sensitivity of downstream detection methods such as ddPCR, sequencing, and clamped enzymatic amplification. We demonstrated 60-fold enrichment of the cancer biomarker KRAS G12D and ∼100-fold increased sensitivity of Peptide Nucleic Acid (PNA) and Xenonucleic Acid (XNA) clamp PCR, enabling detection of low-frequency (<0.01%) mutant alleles (∼1 copy) in blood samples of pancreatic cancer patients. NAVIGATER surpasses Cas9-based assays (e.g. DASH, Depletion of Abundant Sequences by Hybridization), identifying more mutation-positive samples when combined with XNA-PCR. Moreover, TtAgo does not require targets to contain any specific protospacer-adjacent motifs (PAM); is a multi-turnover enzyme; cleaves ssDNA, dsDNA and RNA targets in a single assay; and operates at elevated temperatures, providing high selectivity and compatibility with polymerases.


Sujet(s)
Protéines Argonaute/génétique , Acides nucléiques acellulaires/génétique , Tumeurs/génétique , Acides nucléiques peptidiques/génétique , Allèles , Humains , Mutation/génétique , Tumeurs/diagnostic , Tumeurs/anatomopathologie , Acides nucléiques peptidiques/isolement et purification , Thermus thermophilus/génétique
10.
Cytometry A ; 93(12): 1226-1233, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30549400

RÉSUMÉ

Circulating tumor cells (CTCs) carry valuable biological information. While enumeration of CTCs in peripheral blood is an FDA-approved prognostic indicator of survival in metastatic prostate and other cancers, analysis of CTC phenotypic and genomic markers is needed to identify cancer origin and elucidate pathways that can guide therapeutic selection for personalized medicine. Given the emergence of single-cell mRNA sequencing technologies, a method is needed to isolate CTCs with high sensitivity and specificity as well as compatibility with downstream genomic analysis. Flow cytometry is a powerful tool to analyze and sort single cells, but pre-enrichment is required prior to flow sorting for efficient isolation of CTCs due to the extreme low frequency of CTCs in blood (one in billions of blood cells). While current enrichment technologies often require many steps and result in poor recovery, we demonstrate a magnetic separator and acoustic microfluidic focusing chip integrated system that enriches rare cells in-line with FACS™ (fluorescent activated cell sorting) and single-cell sequencing. This system analyzes, isolates, and index sorts single cells directly into 96-well plates containing reagents for Molecular Indexing (MI) and transcriptional profiling of single cells. With an optimized workflow using the integrated enrichment-FACS system, we performed a proof-of-concept experiment with spiked prostate cancer cells in peripheral blood and achieved: (i) a rapid one-step process to isolate rare cancer cells from lysed whole blood; (ii) an average of 92% post-enrichment cancer cell recovery (R2 = 0.9998) as compared with 55% recovery for a traditional benchtop workflow; and (iii) detection of differentially expressed genes at a single cell level that are consistent with reported cell-type dependent expression signatures for prostate cancer cells. These model system results lay the groundwork for applying our approach to human blood samples from prostate and other cancer patients, and support the enrichment-FACS system as a flexible solution for isolation and characterization of CTCs for cancer diagnosis. © 2018 International Society for Advancement of Cytometry.


Sujet(s)
Tumeurs/anatomopathologie , Cellules tumorales circulantes/anatomopathologie , Analyse sur cellule unique/méthodes , Numération cellulaire/méthodes , Lignée cellulaire tumorale , Séparation cellulaire/méthodes , Cytométrie en flux/méthodes , Humains
11.
Cancer Res ; 78(13): 3688-3697, 2018 07 01.
Article de Anglais | MEDLINE | ID: mdl-29735554

RÉSUMÉ

Improved diagnostics for pancreatic ductal adenocarcinoma (PDAC) to detect the disease at earlier, curative stages and to guide treatments is crucial to progress against this disease. The development of a liquid biopsy for PDAC has proven challenging due to the sparsity and variable phenotypic expression of circulating biomarkers. Here we report methods we developed for isolating specific subsets of extracellular vesicles (EV) from plasma using a novel magnetic nanopore capture technique. In addition, we present a workflow for identifying EV miRNA biomarkers using RNA sequencing and machine-learning algorithms, which we used in combination to classify distinct cancer states. Applying this approach to a mouse model of PDAC, we identified a biomarker panel of 11 EV miRNAs that could distinguish mice with PDAC from either healthy mice or those with precancerous lesions in a training set of n = 27 mice and a user-blinded validation set of n = 57 mice (88% accuracy in a three-way classification). These results provide strong proof-of-concept support for the feasibility of using EV miRNA profiling and machine learning for liquid biopsy.Significance: These findings present a panel of extracellular vesicle miRNA blood-based biomarkers that can detect pancreatic cancer at a precancerous stage in a transgenic mouse model. Cancer Res; 78(13); 3688-97. ©2018 AACR.


Sujet(s)
Carcinome du canal pancréatique/diagnostic , MicroARN circulant/génétique , Exosomes/génétique , Tumeurs du pancréas/diagnostic , Animaux , Marqueurs biologiques tumoraux , Carcinome du canal pancréatique/sang , Carcinome du canal pancréatique/génétique , Lignée cellulaire tumorale , MicroARN circulant/isolement et purification , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes/méthodes , Humains , Biopsie liquide/méthodes , Nanoparticules de magnétite , Souris , Souris transgéniques , Nanopores , Tumeurs du pancréas/sang , Tumeurs du pancréas/génétique , Analyse de séquence d'ARN
12.
Sci Rep ; 8(1): 5035, 2018 03 22.
Article de Anglais | MEDLINE | ID: mdl-29568081

RÉSUMÉ

Comprehensive molecular analysis of rare circulating tumor cells (CTCs) and cell clusters is often hampered by low throughput and purity, as well as cell loss. To address this, we developed a fully integrated platform for flow cytometry-based isolation of CTCs and clusters from blood that can be combined with whole transcriptome analysis or targeted RNA transcript quantification. Downstream molecular signature can be linked to cell phenotype through index sorting. This newly developed platform utilizes in-line magnetic particle-based leukocyte depletion, and acoustic cell focusing and washing to achieve >98% reduction of blood cells and non-cellular debris, along with >1.5 log-fold enrichment of spiked tumor cells. We could also detect 1 spiked-in tumor cell in 1 million WBCs in 4/7 replicates. Importantly, the use of a large 200µm nozzle and low sheath pressure (3.5 psi) minimized shear forces, thereby maintaining cell viability and integrity while allowing for simultaneous recovery of single cells and clusters from blood. As proof of principle, we isolated and transcriptionally characterized 63 single CTCs from a genetically engineered pancreatic cancer mouse model (n = 12 mice) and, using index sorting, were able to identify distinct epithelial and mesenchymal sub-populations based on linked single cell protein and gene expression.


Sujet(s)
Cellules tumorales circulantes/métabolisme , Tumeurs du pancréas/anatomopathologie , Analyse sur cellule unique/méthodes , Animaux , Lignée cellulaire tumorale/transplantation , Séparation cellulaire/méthodes , Modèles animaux de maladie humaine , Cytométrie en flux/méthodes , Analyse de profil d'expression de gènes/méthodes , Humains , Techniques de déleucocytation/instrumentation , Techniques de déleucocytation/méthodes , Biopsie liquide/méthodes , Aimants , Souris , Tumeurs du pancréas/sang , Tumeurs du pancréas/génétique
13.
ACS Nano ; 11(11): 11182-11193, 2017 11 28.
Article de Anglais | MEDLINE | ID: mdl-29019651

RÉSUMÉ

Circulating exosomes contain a wealth of proteomic and genetic information, presenting an enormous opportunity in cancer diagnostics. While microfluidic approaches have been used to successfully isolate cells from complex samples, scaling these approaches for exosome isolation has been limited by the low throughput and susceptibility to clogging of nanofluidics. Moreover, the analysis of exosomal biomarkers is confounded by substantial heterogeneity between patients and within a tumor itself. To address these challenges, we developed a multichannel nanofluidic system to analyze crude clinical samples. Using this platform, we isolated exosomes from healthy and diseased murine and clinical cohorts, profiled the RNA cargo inside of these exosomes, and applied a machine learning algorithm to generate predictive panels that could identify samples derived from heterogeneous cancer-bearing individuals. Using this approach, we classified cancer and precancer mice from healthy controls, as well as pancreatic cancer patients from healthy controls, in blinded studies.


Sujet(s)
Exosomes/génétique , Techniques d'analyse microfluidique/méthodes , Tumeurs du pancréas/diagnostic , Protéomique , Animaux , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Exosomes/anatomopathologie , Humains , Apprentissage machine , Souris , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , ARN messager/génétique
14.
Lab Chip ; 17(18): 3086-3096, 2017 09 12.
Article de Anglais | MEDLINE | ID: mdl-28809985

RÉSUMÉ

The use of microtechnology for the highly selective isolation and sensitive detection of circulating tumor cells has shown enormous promise. One challenge for this technology is that the small feature sizes - which are the key to this technology's performance - can result in low sample throughput and susceptibility to clogging. Additionally, conventional molecular analysis of CTCs often requires cells to be taken off-chip for sample preparation and purification before analysis, leading to the loss of rare cells. To address these challenges, we have developed a microchip platform that combines fast, magnetic micropore based negative immunomagnetic selection (>10 mL h-1) with rapid on-chip in situ RNA profiling (>100× faster than conventional RNA labeling). This integrated chip can isolate both rare circulating cells and cell clusters directly from whole blood and allow individual cells to be profiled for multiple RNA cancer biomarkers, achieving sample-to-answer in less than 1 hour for 10 mL of whole blood. To demonstrate the power of this approach, we applied our device to the circulating tumor cell based diagnosis of pancreatic cancer. We used a genetically engineered lineage-labeled mouse model of pancreatic cancer (KPCY) to validate the performance of our chip. We show that in a cohort of patient samples (N = 25) that this device can detect and perform in situ RNA analysis on circulating tumor cells in patients with pancreatic cancer, even in those with extremely sparse CTCs (<1 CTC mL-1 of whole blood).


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Nanoparticules de magnétite/composition chimique , Techniques d'analyse microfluidique/instrumentation , Cellules tumorales circulantes/composition chimique , ARN messager/analyse , Animaux , Marqueurs biologiques tumoraux/métabolisme , Modèles animaux de maladie humaine , Humains , Souris , Techniques d'analyse microfluidique/méthodes , Cellules tumorales circulantes/métabolisme , Tumeurs du pancréas/diagnostic , Tumeurs du pancréas/métabolisme , ARN messager/métabolisme
15.
Adv Exp Med Biol ; 994: 105-118, 2017.
Article de Anglais | MEDLINE | ID: mdl-28560670

RÉSUMÉ

Circulating tumor cells provide a non-invasive source of tumor material that can be valuable at all stages of disease management, including screening and early diagnosis, monitoring response to therapy, identifying therapeutic targets, and assessing development of drug resistance. Cells isolated from the blood of cancer patients can be used for phenotypic analysis, tumor genotyping, transcriptional profiling, as well as for ex vivo culture of isolated cells. There are a variety of novel technologies currently being developed for the detection and analysis of rare cells in circulation of cancer patients. Flow cytometry is a powerful cell analysis platform that is increasingly being used in this field of study due to its relatively high throughput and versatility with respect to the large number of commercially available antibodies and fluorescent probes available to translational and clinical researchers. More importantly, it offers the ability to easily recover viable cells with high purity that are suitable for downstream molecular analysis, thus making it an attractive technology for cancer research and as a diagnostic tool.


Sujet(s)
Marqueurs biologiques tumoraux/sang , Cytométrie en flux/méthodes , Tumeurs/sang , Cellules tumorales circulantes/métabolisme , Humains
16.
Nat Commun ; 7: 12819, 2016 09 15.
Article de Anglais | MEDLINE | ID: mdl-27628423

RÉSUMÉ

Most cancer-associated deaths result from metastasis. However, it remains unknown whether the size, microenvironment or other features of a metastatic lesion dictate its behaviour or determine the efficacy of chemotherapy in the adjuvant (micrometastatic) setting. Here we delineate the natural history of metastasis in an autochthonous model of pancreatic ductal adenocarcinoma (PDAC), using lineage tracing to examine the evolution of disseminated cancer cells and their associated microenvironment. With increasing size, lesions shift from mesenchymal to epithelial histology, become hypovascular and accumulate a desmoplastic stroma, ultimately recapitulating the primary tumours from which they arose. Moreover, treatment with gemcitabine and nab-paclitaxel significantly reduces the overall number of metastases by inducing cell death in lesions of all sizes, challenging the paradigm that PDAC stroma imposes a critical barrier to drug delivery. These results illuminate the cellular dynamics of metastatic progression and suggest that adjuvant chemotherapy affords a survival benefit by directly targeting micrometastases.


Sujet(s)
Carcinome du canal pancréatique/secondaire , Tumeurs du foie/secondaire , Foie/anatomopathologie , Tumeurs du pancréas/anatomopathologie , Microenvironnement tumoral , Sujet âgé , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Carcinome du canal pancréatique/vascularisation , Carcinome du canal pancréatique/traitement médicamenteux , Lignage cellulaire , Femelle , Humains , Foie/effets des médicaments et des substances chimiques , Tumeurs du foie/vascularisation , Tumeurs du foie/traitement médicamenteux , Mâle , Souris , Adulte d'âge moyen , Métastase tumorale , Tumeurs du pancréas/vascularisation , Tumeurs du pancréas/traitement médicamenteux
17.
Blood ; 126(22): 2479-83, 2015 Nov 26.
Article de Anglais | MEDLINE | ID: mdl-26443624

RÉSUMÉ

The development of the dual Janus kinase 1/2 (JAK1/2) inhibitor ruxolitinib for the treatment of myeloproliferative neoplasms (MPNs) has led to studies of ruxolitinib in other clinical contexts, including JAK-mutated acute lymphoblastic leukemia (ALL). However, the limited ability of JAK inhibition to induce molecular or clinicopathological responses in MPNs suggests a need for development of better therapies for JAK kinase-dependent malignancies. Here, we demonstrate that heat shock protein 90 (HSP90) inhibition using a purine-scaffold HSP90 inhibitor in early clinical development is an effective therapeutic approach in JAK-dependent ALL and can overcome persistence to JAK-inhibitor therapy in ALL cells.


Sujet(s)
Benzodioxoles/pharmacologie , Protéines du choc thermique HSP90 , Janus kinase 1 , Kinase Janus-2 , Protéines tumorales , Leucémie-lymphome lymphoblastique à précurseurs B et T , Purines/pharmacologie , Animaux , Femelle , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/génétique , Protéines du choc thermique HSP90/métabolisme , Humains , Janus kinase 1/génétique , Janus kinase 1/métabolisme , Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Mâle , Souris , Mutation , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Cancer Discov ; 5(3): 316-31, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25572172

RÉSUMÉ

UNLABELLED: The identification of JAK2/MPL mutations in patients with myeloproliferative neoplasms (MPN) has led to the clinical development of JAK kinase inhibitors, including ruxolitinib. Ruxolitinib reduces splenomegaly and systemic symptoms in myelofibrosis and improves overall survival; however, the mechanism by which JAK inhibitors achieve efficacy has not been delineated. Patients with MPN present with increased levels of circulating proinflammatory cytokines, which are mitigated by JAK inhibitor therapy. We sought to elucidate mechanisms by which JAK inhibitors attenuate cytokine-mediated pathophysiology. Single-cell profiling demonstrated that hematopoietic cells from myelofibrosis models and patient samples aberrantly secrete inflammatory cytokines. Pan-hematopoietic Stat3 deletion reduced disease severity and attenuated cytokine secretion, with similar efficacy as observed with ruxolitinib therapy. In contrast, Stat3 deletion restricted to MPN cells did not reduce disease severity or cytokine production. Consistent with these observations, we found that malignant and nonmalignant cells aberrantly secrete cytokines and JAK inhibition reduces cytokine production from both populations. SIGNIFICANCE: Our results demonstrate that JAK-STAT3-mediated cytokine production from malignant and nonmalignant cells contributes to MPN pathogenesis and that JAK inhibition in both populations is required for therapeutic efficacy. These findings provide novel insight into the mechanisms by which JAK kinase inhibition achieves therapeutic efficacy in MPNs.


Sujet(s)
Transformation cellulaire néoplasique/métabolisme , Janus kinases/métabolisme , Syndromes myéloprolifératifs/métabolisme , Facteurs de transcription STAT/métabolisme , Transduction du signal , Animaux , Antinéoplasiques/pharmacologie , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Cytokines/métabolisme , Modèles animaux de maladie humaine , Délétion de gène , Humains , Médiateurs de l'inflammation/métabolisme , Janus kinase 1/antagonistes et inhibiteurs , Kinase Janus-2/antagonistes et inhibiteurs , Janus kinases/génétique , Antigènes CD45/génétique , Antigènes CD45/métabolisme , Souris , Souris knockout , Mutation , Cellules myéloïdes/effets des médicaments et des substances chimiques , Cellules myéloïdes/métabolisme , Syndromes myéloprolifératifs/traitement médicamenteux , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/anatomopathologie , Myélofibrose primitive/génétique , Myélofibrose primitive/métabolisme , Myélofibrose primitive/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Facteurs de transcription STAT/génétique , Transduction du signal/effets des médicaments et des substances chimiques
19.
Blood ; 124(14): 2280-4, 2014 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-25115888

RÉSUMÉ

JAK inhibitor treatment is limited by the variable development of anemia and thrombocytopenia thought to be due to on-target JAK2 inhibition. We evaluated the impact of Jak2 deletion in platelets (PLTs) and megakaryocytes (MKs) on blood counts, stem/progenitor cells, and Jak-Stat signaling. Pf4-Cre-mediated Jak2 deletion in PLTs and MKs did not compromise PLT formation but caused thrombocytosis, and resulted in expansion of MK progenitors and Lin(-)Sca1(+)Kit+ cells. Serum thrombopoietin (TPO) was maintained at normal levels in Pf4-Cre-positive Jak2(f/f) mice, consistent with reduced internalization/turnover by Jak2-deficient PLTs. These data demonstrate that Jak2 in terminal megakaryopoiesis is not required for PLT production, and that Jak2 loss in PLTs and MKs results in non-autonomous expansion of stem/progenitors and of MKs and PLTs via dysregulated TPO turnover. This suggests that the thrombocytopenia frequently seen with JAK inhibitor treatment is not due to JAK2 inhibition in PLTs and MKs, but rather due to JAK2 inhibition in stem/progenitor cells.


Sujet(s)
Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Thrombocytose/métabolisme , Thrombopoïèse/physiologie , Animaux , Plaquettes/cytologie , Croisements génétiques , Délétion de gène , Régulation de l'expression des gènes codant pour des enzymes , Cellules souches hématopoïétiques/cytologie , Mégacaryocytes/cytologie , Souris , Transduction du signal , Cellules souches/cytologie , Thrombopoïétine/sang , Thrombopoïétine/métabolisme
20.
Blood ; 123(13): 2075-83, 2014 Mar 27.
Article de Anglais | MEDLINE | ID: mdl-24470592

RÉSUMÉ

The discovery of JAK2/MPL mutations in patients with myeloproliferative neoplasms (MPN) led to clinical development of Janus kinase (JAK) inhibitors for treatment of MPN. These inhibitors improve constitutional symptoms and splenomegaly but do not significantly reduce mutant allele burden in patients. We recently showed that chronic exposure to JAK inhibitors results in inhibitor persistence via JAK2 transactivation and persistent JAK-signal transducer and activator of transcription signaling. We performed genetic and pharmacologic studies to determine whether improved JAK2 inhibition would show increased efficacy in MPN models and primary samples. Jak2 deletion in vivo led to profound reduction in disease burden not seen with JAK inhibitors, and deletion of Jak2 following chronic ruxolitinib therapy markedly reduced mutant allele burden. This demonstrates that JAK2 remains an essential target in MPN cells that survive in the setting of chronic JAK inhibition. Combination therapy with the heat shock protein 90 (HSP90) inhibitor PU-H71 and ruxolitinib reduced total and phospho-JAK2 and achieved more potent inhibition of downstream signaling than ruxolitinib monotherapy. Combination treatment improved blood counts, spleen weights, and reduced bone marrow fibrosis compared with ruxolitinib alone. These data suggest alternate approaches that increase JAK2 targeting, including combination JAK/HSP90 inhibitor therapy, are warranted in the clinical setting.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Thérapie génétique/méthodes , Kinase Janus-2/génétique , Thérapie moléculaire ciblée/méthodes , Syndromes myéloprolifératifs/traitement médicamenteux , Substitution d'acide aminé , Animaux , Tumeurs de la moelle osseuse/traitement médicamenteux , Transformation cellulaire néoplasique/génétique , Association thérapeutique , Délétion de gène , Kinase Janus-2/antagonistes et inhibiteurs , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris transgéniques , Récepteurs à la thrombopoïétine/génétique , Résultat thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...