Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
1.
Clin Cancer Res ; 24(2): 360-369, 2018 01 15.
Article de Anglais | MEDLINE | ID: mdl-29118061

RÉSUMÉ

Purpose: Small-cell lung cancer (SCLC) is an often-fatal neuroendocrine carcinoma usually presenting as extensive disease, carrying a 3% 5-year survival. Despite notable advances in SCLC genomics, new therapies remain elusive, largely due to a lack of druggable targets.Experimental Design: We used a high-throughput drug screen to identify a venetoclax-sensitive SCLC subpopulation and validated the findings with multiple patient-derived xenografts of SCLC.Results: Our drug screen consisting of a very large collection of cell lines demonstrated that venetoclax, an FDA-approved BCL-2 inhibitor, was found to be active in a substantial fraction of SCLC cell lines. Venetoclax induced BIM-dependent apoptosis in vitro and blocked tumor growth and induced tumor regressions in mice bearing high BCL-2-expressing SCLC tumors in vivo BCL-2 expression was a predictive biomarker for sensitivity in SCLC cell lines and was highly expressed in a subset of SCLC cell lines and tumors, suggesting that a substantial fraction of patients with SCLC could benefit from venetoclax. Mechanistically, we uncover a novel role for gene methylation that helped discriminate high BCL-2-expressing SCLCs.Conclusions: Altogether, our findings identify venetoclax as a promising new therapy for high BCL-2-expressing SCLCs. Clin Cancer Res; 24(2); 360-9. ©2017 AACR.


Sujet(s)
Antinéoplasiques/pharmacologie , Composés hétérocycliques bicycliques/pharmacologie , Expression des gènes , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Protéines proto-oncogènes c-bcl-2/antagonistes et inhibiteurs , Protéines proto-oncogènes c-bcl-2/génétique , Carcinome pulmonaire à petites cellules/génétique , Sulfonamides/pharmacologie , Animaux , Antinéoplasiques/usage thérapeutique , Apoptose , Composés hétérocycliques bicycliques/usage thérapeutique , Lignée cellulaire tumorale , Méthylation de l'ADN , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/génétique , Régulation de l'expression des gènes tumoraux/génétique , Humains , Souris , Régions promotrices (génétique) , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/anatomopathologie , Sulfonamides/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Oncotarget ; 8(43): 73419-73432, 2017 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-29088717

RÉSUMÉ

Small cell lung cancer (SCLC) is a recalcitrant cancer for which no new treatments have been approved in over 30 years. While molecular subtyping now guides treatment selection for patients with non-small cell lung cancer and other cancers, SCLC is still treated as a single disease entity. Using model-based clustering, we found two major proteomic subtypes of SCLC characterized by either high thyroid transcription factor-1 (TTF1)/low cMYC protein expression or high cMYC/low TTF1. Applying "drug target constellation" (DTECT) mapping, we further show that protein levels of TTF1 and cMYC predict response to targeted therapies including aurora kinase, Bcl2, and HSP90 inhibitors. Levels of TTF1 and DLL3 were also highly correlated in preclinical models and patient tumors. TTF1 (used in the diagnosis lung cancer) could therefore be used as a surrogate of DLL3 expression to identify patients who may respond to the DLL3 antibody-drug conjugate rovalpituzumab tesirine. These findings suggest that TTF1, cMYC or other protein markers identified here could be used to identify subgroups of SCLC patients who may respond preferentially to several emerging targeted therapies.

3.
Cancer Res ; 77(14): 3931-3941, 2017 07 15.
Article de Anglais | MEDLINE | ID: mdl-28487384

RÉSUMÉ

The Notch ligand DLL3 has emerged as a novel therapeutic target expressed in small cell lung cancer (SCLC) and high-grade neuroendocrine carcinomas. Rovalpituzumab teserine (Rova-T; SC16LD6.5) is a first-in-class DLL3-targeted antibody-drug conjugate with encouraging initial safety and efficacy profiles in SCLC in the clinic. Here we demonstrate that tumor expression of DLL3, although orders of magnitude lower in surface protein expression than typical oncology targets of immunoPET, can serve as an imaging biomarker for SCLC. We developed 89Zr-labeled SC16 antibody as a companion diagnostic agent to facilitate selection of patients for treatment with Rova-T based on a noninvasive interrogation of the in vivo status of DLL3 expression using PET imaging. Despite low cell-surface abundance of DLL3, immunoPET imaging with 89Zr-labeled SC16 antibody enabled delineation of subcutaneous and orthotopic SCLC tumor xenografts as well as distant organ metastases with high sensitivity. Uptake of the radiotracer in tumors was concordant with levels of DLL3 expression and, most notably, DLL3 immunoPET yielded rank-order correlation for response to SC16LD6.5 therapy in SCLC patient-derived xenograft models. Cancer Res; 77(14); 3931-41. ©2017 AACR.


Sujet(s)
Protéines et peptides de signalisation intracellulaire/biosynthèse , Tumeurs du poumon/métabolisme , Protéines membranaires/biosynthèse , Carcinome pulmonaire à petites cellules/métabolisme , Cellules A549 , Animaux , Lignée cellulaire tumorale , Femelle , Hétérogreffes , Humains , Immunoconjugués , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/immunologie , Tumeurs du poumon/imagerie diagnostique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Protéines membranaires/génétique , Protéines membranaires/immunologie , Souris , Souris nude , Métastase tumorale , Tomographie par émission de positons , Carcinome pulmonaire à petites cellules/imagerie diagnostique , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/anatomopathologie
4.
Sci Transl Med ; 9(372)2017 01 11.
Article de Anglais | MEDLINE | ID: mdl-28077676

RÉSUMÉ

Disease relapse after treatment is common in triple-negative breast cancer (TNBC), ovarian cancer (OVCA), and non-small cell lung cancer (NSCLC). Therapies that target tumor-initiating cells (TICs) should improve patient survival by eliminating the cells that can drive tumor recurrence and metastasis. We demonstrate that protein tyrosine kinase 7 (PTK7), a highly conserved but catalytically inactive receptor tyrosine kinase in the Wnt signaling pathway, is enriched on TICs in low-passage TNBC, OVCA, and NSCLC patient-derived xenografts (PDXs). To deliver a potent anticancer drug to PTK7-expressing TICs, we generated a targeted antibody-drug conjugate (ADC) composed of a humanized anti-PTK7 monoclonal antibody, a cleavable valine-citrulline-based linker, and Aur0101, an auristatin microtubule inhibitor. The PTK7-targeted ADC induced sustained tumor regressions and outperformed standard-of-care chemotherapy. Moreover, the ADC specifically reduced the frequency of TICs, as determined by serial transplantation experiments. In addition to reducing the TIC frequency, the PTK7-targeted ADC may have additional antitumor mechanisms of action, including the inhibition of angiogenesis and the stimulation of immune cells. Together, these preclinical data demonstrate the potential for the PTK7-targeted ADC to improve the long-term survival of cancer patients.


Sujet(s)
Anticorps/usage thérapeutique , Molécules d'adhérence cellulaire/composition chimique , Immunoconjugués/usage thérapeutique , Cellules souches tumorales/effets des médicaments et des substances chimiques , Récepteurs à activité tyrosine kinase/composition chimique , Aminobenzoates/usage thérapeutique , Animaux , Antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/thérapie , Molécules d'adhérence cellulaire/immunologie , Lignée cellulaire tumorale , Essais cliniques comme sujet , Femelle , Humains , Immunothérapie/méthodes , Tumeurs du poumon/immunologie , Tumeurs du poumon/thérapie , Macaca fascicularis , Souris , Souris de lignée NOD , Souris SCID , Microtubules/composition chimique , Récidive tumorale locale/traitement médicamenteux , Oligopeptides/usage thérapeutique , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/thérapie , Récepteurs à activité tyrosine kinase/immunologie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/thérapie , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Lancet Oncol ; 18(1): 42-51, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27932068

RÉSUMÉ

BACKGROUND: Rovalpituzumab tesirine is a first-in-class antibody-drug conjugate directed against delta-like protein 3 (DLL3), a novel target identified in tumour-initiating cells and expressed in more than 80% of patients with small-cell lung cancer. We aimed to assess the safety and activity of rovalpituzumab tesirine in patients who progressed after one or more previous regimen. METHODS: We conducted a phase 1 open-label study at ten cancer centres in the USA. Eligible patients were aged 18 years or older and had histologically or cytologically confirmed small-cell lung cancer or large-cell neuroendocrine tumours with progressive measurable disease (according to Response Evaluation Criteria in Solid Tumors [RECIST], version 1.1) previously treated with one or two chemotherapeutic regimens, including a platinum-based regimen. We assigned patients to dose-escalation or expansion cohorts, ranging from 0·05 mg/kg to 0·8 mg/kg rovalpituzumab tesirine intravenously every 3 weeks or every 6 weeks, followed by investigation of the dose schedules 0·3 mg/kg and 0·4 mg/kg every 6 weeks and 0·2 mg/kg every 3 weeks. Primary objectives were to assess the safety of rovalpituzumab tesirine, including the maximum tolerated dose and dose-limiting toxic effects. The primary activity endpoint was objective response by intention-to-treat analysis. This study is registered with ClinicalTrials.gov, number NCT01901653. The study is closed to enrolment; this report focuses on the cohort with small-cell lung cancer. FINDINGS: Between July 22, 2013, and Aug 10, 2015, 82 patients were enrolled, including 74 patients with small-cell lung cancer and eight with large-cell neuroendocrine carcinoma, all of whom received at least one dose of rovalpituzumab tesirine. Dose-limiting toxic effects of rovalpituzumab tesirine occurred at a dose of 0·8 mg/kg every 3 weeks, including grade 4 thrombocytopenia (in two of two patients at that dose level) and grade 4 liver function test abnormalities (in one patient). The most frequent grade 3 or worse treatment-related adverse events in 74 patients with small-cell lung cancer were thrombocytopenia (eight [11%]), pleural effusion (six [8%]), and increased lipase (five [7%]). Drug-related serious adverse events occurred in 28 (38%) of 74 patients. The maximum tolerated dose of rovalpituzumab tesirine was 0·4 mg/kg every 3 weeks; the recommended phase 2 dose and schedule is 0·3 mg/kg every 6 weeks. At active doses of rovalpituzumab tesirine (0·2 mg/kg or 0·4 mg/kg every 3 weeks or 0·3 mg/kg or 0·4 mg/kg every 6 weeks), 11 (18%) of 60 assessable patients had a confirmed objective response. 11 (18%) of 60 assessable patients had a confirmed objective response, including ten (38%) of 26 patients confirmed to have high DLL3 expression (expression in 50% or more of tumour cells). INTERPRETATION: Rovalpituzumab tesirine shows encouraging single-agent antitumour activity with a manageable safety profile. Further development of rovalpituzumab tesirine in DLL3-expressing malignant diseases is warranted. FUNDING: Stemcentrx Inc.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Benzodiazépinones/usage thérapeutique , Carcinome à grandes cellules/traitement médicamenteux , Carcinome neuroendocrine/traitement médicamenteux , Immunoconjugués/usage thérapeutique , Protéines et peptides de signalisation intracellulaire/immunologie , Tumeurs du poumon/traitement médicamenteux , Protéines membranaires/immunologie , Récidive tumorale locale/traitement médicamenteux , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Sujet âgé , Carcinome à grandes cellules/immunologie , Carcinome à grandes cellules/anatomopathologie , Carcinome neuroendocrine/immunologie , Carcinome neuroendocrine/anatomopathologie , Relation dose-effet des médicaments , Femelle , Études de suivi , Humains , Immunoconjugués/pharmacologie , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Récidive tumorale locale/immunologie , Récidive tumorale locale/anatomopathologie , Stadification tumorale , Pronostic , Carcinome pulmonaire à petites cellules/immunologie , Carcinome pulmonaire à petites cellules/anatomopathologie , Taux de survie
6.
Cell Rep ; 16(3): 644-56, 2016 07 19.
Article de Anglais | MEDLINE | ID: mdl-27373157

RÉSUMÉ

Small cell lung cancer (SCLC) is a neuroendocrine lung cancer characterized by fast growth, early dissemination, and rapid resistance to chemotherapy. We identified a population of long-term tumor-propagating cells (TPCs) in a mouse model of SCLC. This population, marked by high levels of EpCAM and CD24, is also prevalent in human primary SCLC tumors. Murine SCLC TPCs are numerous and highly proliferative but not intrinsically chemoresistant, indicating that not all clinical features of SCLC are linked to TPCs. SCLC TPCs possess a distinct transcriptional profile compared to non-TPCs, including elevated MYC activity. Genetic and pharmacological inhibition of MYC in SCLC cells to non-TPC levels inhibits long-term propagation but not short-term growth. These studies identify a highly tumorigenic population of SCLC cells in mouse models, cell lines, and patient tumors and a means to target them in this most fatal form of lung cancer.


Sujet(s)
Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire à petites cellules/anatomopathologie , Animaux , Carcinogenèse/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/physiologie , Modèles animaux de maladie humaine , Humains , Tumeurs du poumon/génétique , Souris , Carcinome pulmonaire à petites cellules/génétique , Transcription génétique/physiologie
7.
Mol Cell Oncol ; 3(2): e1101515, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-27308627

RÉSUMÉ

Delta-like protein 3 (DLL3) is a novel and tractable tumor-initiating cell-associated target for the antibody-drug conjugate SC16LD6.5 in high-grade pulmonary neuroendocrine tumors. Elevated expression of DLL3, an inhibitor of Notch pathway activation, marks the second recent observation that impairment of Notch receptor signaling may play a critical role in neuroendocrine tumorigenesis.

8.
Sci Transl Med ; 7(302): 302ra136, 2015 Aug 26.
Article de Anglais | MEDLINE | ID: mdl-26311731

RÉSUMÉ

The high-grade pulmonary neuroendocrine tumors, small cell lung cancer (SCLC) and large cell neuroendocrine carcinoma (LCNEC), remain among the most deadly malignancies. Therapies that effectively target and kill tumor-initiating cells (TICs) in these cancers should translate to improved patient survival. Patient-derived xenograft (PDX) tumors serve as excellent models to study tumor biology and characterize TICs. Increased expression of delta-like 3 (DLL3) was discovered in SCLC and LCNEC PDX tumors and confirmed in primary SCLC and LCNEC tumors. DLL3 protein is expressed on the surface of tumor cells but not in normal adult tissues. A DLL3-targeted antibody-drug conjugate (ADC), SC16LD6.5, comprised of a humanized anti-DLL3 monoclonal antibody conjugated to a DNA-damaging pyrrolobenzodiazepine (PBD) dimer toxin, induced durable tumor regression in vivo across multiple PDX models. Serial transplantation experiments executed with limiting dilutions of cells provided functional evidence confirming that the lack of tumor recurrence after SC16LD6.5 exposure resulted from effective targeting of DLL3-expressing TICs. In vivo efficacy correlated with DLL3 expression, and responses were observed in PDX models initiated from patients with both limited and extensive-stage disease and were independent of their sensitivity to standard-of-care chemotherapy regimens. SC16LD6.5 effectively targets and eradicates DLL3-expressing TICs in SCLC and LCNEC PDX tumors and is a promising first-in-class ADC for the treatment of high-grade pulmonary neuroendocrine tumors.


Sujet(s)
Anticorps monoclonaux/immunologie , Antinéoplasiques/usage thérapeutique , Immunoconjugués/usage thérapeutique , Protéines et peptides de signalisation intracellulaire/immunologie , Tumeurs du poumon/traitement médicamenteux , Protéines membranaires/immunologie , Tumeurs neuroendocrines/traitement médicamenteux , Animaux , Femelle , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Tumeurs du poumon/métabolisme , Protéines membranaires/métabolisme , Souris , Souris de lignée NOD , Souris SCID , Tumeurs neuroendocrines/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
9.
PLoS One ; 10(5): e0125255, 2015.
Article de Anglais | MEDLINE | ID: mdl-25955027

RÉSUMÉ

Small cell lung cancer (SCLC) is a devastating disease with limited treatment options. Due to its early metastatic nature and rapid growth, surgical resection is rare. Standard of care treatment regimens remain largely unchanged since the 1980's, and five-year survival lingers near 5%. Patient-derived xenograft (PDX) models have been established for other tumor types, amplifying material for research and serving as models for preclinical experimentation; however, limited availability of primary tissue has curtailed development of these models for SCLC. The objective of this study was to establish PDX models from commonly collected fine needle aspirate biopsies of primary SCLC tumors, and to assess their utility as research models of primary SCLC tumors. These transbronchial needle aspirates efficiently engrafted as xenografts, and tumor histomorphology was similar to primary tumors. Resulting tumors were further characterized by H&E and immunohistochemistry, cryopreserved, and used to propagate tumor-bearing mice for the evaluation of standard of care chemotherapy regimens, to assess their utility as models for tumors in SCLC patients. When treated with Cisplatin and Etoposide, tumor-bearing mice responded similarly to patients from whom the tumors originated. Here, we demonstrate that PDX tumor models can be efficiently established from primary SCLC transbronchial needle aspirates, even after overnight shipping, and that resulting xenograft tumors are similar to matched primary tumors in cancer patients by both histology and chemo-sensitivity. This method enables physicians at non-research institutions to collaboratively contribute to the rapid establishment of extensive PDX collections of SCLC, enabling experimentation with clinically relevant tissues and development of improved therapies for SCLC patients.


Sujet(s)
Bronches/imagerie diagnostique , Bronches/anatomopathologie , Tumeurs du poumon/imagerie diagnostique , Tumeurs du poumon/anatomopathologie , Carcinome pulmonaire à petites cellules/imagerie diagnostique , Carcinome pulmonaire à petites cellules/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Antigènes néoplasiques/immunologie , Marqueurs biologiques tumoraux/métabolisme , Cytoponction , Humains , Immunohistochimie , Souris , Adulte d'âge moyen , Résultat thérapeutique , Échographie
10.
Clin Cancer Res ; 21(18): 4165-73, 2015 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-26015513

RÉSUMÉ

PURPOSE: Triple-negative breast cancer (TNBC) and ovarian cancer each comprise heterogeneous tumors, for which current therapies have little clinical benefit. Novel therapies that target and eradicate tumor-initiating cells (TIC) are needed to significantly improve survival. EXPERIMENTAL DESIGN: A panel of well-annotated patient-derived xenografts (PDX) was established, and surface markers that enriched for TIC in specific tumor subtypes were empirically determined. The TICs were queried for overexpressed antigens, one of which was selected to be the target of an antibody-drug conjugate (ADC). The efficacy of the ADC was evaluated in 15 PDX models to generate hypotheses for patient stratification. RESULTS: We herein identified E-cadherin (CD324) as a surface antigen able to reproducibly enrich for TIC in well-annotated, low-passage TNBC and ovarian cancer PDXs. Gene expression analysis of TIC led to the identification of Ephrin-A4 (EFNA4) as a prospective therapeutic target. An ADC comprising a humanized anti-EFNA4 monoclonal antibody conjugated to the DNA-damaging agent calicheamicin achieved sustained tumor regressions in both TNBC and ovarian cancer PDX in vivo. Non-claudin low TNBC tumors exhibited higher expression and more robust responses than other breast cancer subtypes, suggesting a specific translational application for tumor subclassification. CONCLUSIONS: These findings demonstrate the potential of PF-06647263 (anti-EFNA4-ADC) as a first-in-class compound designed to eradicate TIC. The use of well-annotated PDX for drug discovery enabled the identification of a novel TIC target, pharmacologic evaluation of the compound, and translational studies to inform clinical development.


Sujet(s)
Aminosides/composition chimique , Anticorps monoclonaux d'origine murine/composition chimique , Ènediynes/composition chimique , Éphrine A4/composition chimique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs du sein triple-négatives/traitement médicamenteux , Animaux , Anticorps monoclonaux humanisés/composition chimique , Antigènes néoplasiques/composition chimique , Lignée cellulaire tumorale , ADN/composition chimique , Conception de médicament , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Cellules HEK293 , Humains , Souris , Souris de lignée NOD , Souris SCID , Cellules souches tumorales/métabolisme , Études prospectives , Répartition aléatoire , Résultat thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Lab Invest ; 93(9): 970-82, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23917877

RÉSUMÉ

Cancer is a heterogeneous disease manifest in many forms. Tumor histopathology can differ significantly among patients and cellular heterogeneity within tumors is common. A primary goal of cancer biologists is to better understand tumorigenesis and cancer progression; however, the complex nature of tumors has posed a substantial challenge to unlocking cancer's secrets. The cancer stem cell (CSC) paradigm for the pathobiology of solid tumors appropriately acknowledges phenotypic and functional tumor cell heterogeneity observed in solid tumors and accounts for the disconnect between drug approval based on response and the general inability of approved therapies to meaningfully impact survival due to their failure to eradicate these most important of cellular targets. First proposed to exist decades ago, CSC have only recently begun to be precisely identified due to technical advancements that facilitate identification, isolation, and interrogation of distinct tumor cell subpopulations with differing ability to form and perpetuate tumors. Precise identification of CSC populations and the complete hierarchy of cells within solid tumors will facilitate more accurate characterization of patient subtypes and ultimately contribute to more personalized and effective therapies. Rapid advancement in the understanding of tumor biology as it exists in patients requires cooperation among institutions, surgeons, pathologists, cancer biologists and patients alike, primarily because this translational research is best done with patient-derived tissue grown in the xenograft setting as patient-derived xenografts. This review calls for a broader change in the approaches taken to study cancer pathobiology, highlights what implications the CSC paradigm has for pathologists and cancer biologists alike, and calls for greater collaboration between institutions, physicians and scientists in order to more rapidly advance our collective understanding of cancer.


Sujet(s)
Transplantation tumorale/méthodes , Tumeurs/anatomopathologie , Cellules souches tumorales/anatomopathologie , Transplantation hétérologue/anatomopathologie , Animaux , Humains , Oncologie médicale
12.
J Thorac Cardiovasc Surg ; 136(4): 1028-1037.e1, 2008 Oct.
Article de Anglais | MEDLINE | ID: mdl-18954646

RÉSUMÉ

OBJECTIVE: Mouse embryonic stem cells have demonstrated potential to restore infarcted myocardium after acute myocardial infarction. Although the underlying mechanism remains controversial, magnetic resonance imaging has provided reliable in vivo assessment of functional recovery after cellular transplants. Multimodal comparison of the restorative effects of mouse embryonic stem cells and mouse embryonic fibroblasts was performed to validate magnetic resonance imaging data and provide mechanistic insight. METHODS: SCID-beige mice (n = 55) underwent coronary artery ligation followed by injection of 2.5 x 10(5) mouse embryonic stem cells, 2.5 x 10(5) mouse embryonic fibroblasts, or normal saline solution. In vivo magnetic resonance imaging of myocardial restoration by mouse embryonic stem cells was evaluated by (1) in vivo pressure-volume loops, (2) in vivo bioluminescence imaging, and (3) ex vivo TaqMan (Roche Molecular Diagnostics, Pleasanton, Calif) polymerase chain reaction and immunohistologic examination. RESULTS: In vivo magnetic resonance imaging demonstrated significant improvement in left ventricular ejection fraction at 1 week in the mouse embryonic stem cell group. This finding was validated with (1) pressure-volume loop analysis demonstrating significantly improved systolic and diastolic functions, (2) bioluminescence imaging and polymerase chain reaction showing superior posttransplant survival of mouse embryonic stem cells, (3) immunohistologic identification of cardiac phenotype within engrafted mouse embryonic stem cells, and (4) polymerase chain reaction measuring increased expressions of angiogenic and antiapoptotic genes and decreased expressions of antifibrotic genes. CONCLUSION: This study validates in vivo magnetic resonance imaging as an effective means of evaluating the restorative potential of mouse embryonic stem cells.


Sujet(s)
Cellules souches embryonnaires/transplantation , Imagerie par résonance magnétique/méthodes , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/chirurgie , Transplantation de cellules souches/méthodes , Remodelage ventriculaire/physiologie , Animaux , Modèles animaux de maladie humaine , Cellules souches embryonnaires/anatomopathologie , Femelle , Fibroblastes/transplantation , Rejet du greffon , Survie du greffon , Immunohistochimie , Souris , Souris SCID , Contraction myocardique/physiologie , Myocytes cardiaques/anatomopathologie , Répartition aléatoire , Reproductibilité des résultats , Sensibilité et spécificité , Débit systolique
13.
PLoS One ; 3(6): e2428, 2008 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-18560594

RÉSUMÉ

BACKGROUND: Patients generally die of cancer after the failure of current therapies to eliminate residual disease. A subpopulation of tumor cells, termed cancer stem cells (CSC), appears uniquely able to fuel the growth of phenotypically and histologically diverse tumors. It has been proposed, therefore, that failure to effectively treat cancer may in part be due to preferential resistance of these CSC to chemotherapeutic agents. The subpopulation of human colorectal tumor cells with an ESA(+)CD44(+) phenotype are uniquely responsible for tumorigenesis and have the capacity to generate heterogeneous tumors in a xenograft setting (i.e. CoCSC). We hypothesized that if non-tumorigenic cells are more susceptible to chemotherapeutic agents, then residual tumors might be expected to contain a higher frequency of CoCSC. METHODS AND FINDINGS: Xenogeneic tumors initiated with CoCSC were allowed to reach approximately 400 mm(3), at which point mice were randomized and chemotherapeutic regimens involving cyclophosphamide or Irinotecan were initiated. Data from individual tumor phenotypic analysis and serial transplants performed in limiting dilution show that residual tumors are enriched for cells with the CoCSC phenotype and have increased tumorigenic cell frequency. Moreover, the inherent ability of residual CoCSC to generate tumors appears preserved. Aldehyde dehydrogenase 1 gene expression and enzymatic activity are elevated in CoCSC and using an in vitro culture system that maintains CoCSC as demonstrated by serial transplants and lentiviral marking of single cell-derived clones, we further show that ALDH1 enzymatic activity is a major mediator of resistance to cyclophosphamide: a classical chemotherapeutic agent. CONCLUSIONS: CoCSC are enriched in colon tumors following chemotherapy and remain capable of rapidly regenerating tumors from which they originated. By focusing on the biology of CoCSC, major resistance mechanisms to specific chemotherapeutic agents can be attributed to specific genes, thereby suggesting avenues for improving cancer therapy.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Camptothécine/analogues et dérivés , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Cyclophosphamide/usage thérapeutique , Cellules souches tumorales/cytologie , Aldehyde dehydrogenase/génétique , Animaux , Camptothécine/usage thérapeutique , Humains , Irinotécan , Souris
14.
Proc Natl Acad Sci U S A ; 104(24): 10158-63, 2007 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-17548814

RÉSUMÉ

Recent observations indicate that, in several types of human cancer, only a phenotypic subset of cancer cells within each tumor is capable of initiating tumor growth. This functional subset of cancer cells is operationally defined as the "cancer stem cell" (CSC) subset. Here we developed a CSC model for the study of human colorectal cancer (CRC). Solid CRC tissues, either primary tissues collected from surgical specimens or xenografts established in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice, were disaggregated into single-cell suspensions and analyzed by flow cytometry. Surface markers that displayed intratumor heterogeneous expression among epithelial cancer cells were selected for cell sorting and tumorigenicity experiments. Individual phenotypic cancer cell subsets were purified, and their tumor-initiating properties were investigated by injection in NOD/SCID mice. Our observations indicate that, in six of six human CRC tested, the ability to engraft in vivo in immunodeficient mice was restricted to a minority subpopulation of epithelial cell adhesion molecule (EpCAM)(high)/CD44+ epithelial cells. Tumors originated from EpCAM(high)/CD44+ cells maintained a differentiated phenotype and reproduced the full morphologic and phenotypic heterogeneity of their parental lesions. Analysis of the surface molecule repertoire of EpCAM(high)/CD44+ cells led to the identification of CD166 as an additional differentially expressed marker, useful for CSC isolation in three of three CRC tested. These results validate the stem cell working model in human CRC and provide a highly robust surface marker profile for CRC stem cell isolation.


Sujet(s)
Antigènes néoplasiques , Marqueurs biologiques tumoraux/immunologie , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Cellules souches/immunologie , Molécule d'adhérence cellulaire des leucocytes activés/immunologie , Animaux , Antigènes néoplasiques/immunologie , Séparation cellulaire , Tumeurs colorectales/génétique , Tumeurs colorectales/chirurgie , Cellules épithéliales/immunologie , Cytométrie en flux , Humains , Antigènes CD44/immunologie , Souris , Souris de lignée NOD , Souris SCID , Transplantation tumorale , Phénotype , Transplantation hétérologue , Test clonogénique de cellules souches tumorales
15.
J Exp Med ; 204(1): 129-39, 2007 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-17227908

RÉSUMÉ

For decades, in vitro expansion of transplantable hematopoietic stem cells (HSCs) has been an elusive goal. Here, we demonstrate that multipotent adult progenitor cells (MAPCs), isolated from green fluorescent protein (GFP)-transgenic mice and expanded in vitro for >40-80 population doublings, are capable of multilineage hematopoietic engraftment of immunodeficient mice. Among MAPC-derived GFP+CD45.2+ cells in the bone marrow of engrafted mice, HSCs were present that could radioprotect and reconstitute multilineage hematopoiesis in secondary and tertiary recipients, as well as myeloid and lymphoid hematopoietic progenitor subsets and functional GFP+ MAPC-derived lymphocytes that were functional. Although hematopoietic contribution by MAPCs was comparable to control KTLS HSCs, approximately 10(3)-fold more MAPCs were required for efficient engraftment. Because GFP+ host-derived CD45.1+ cells were not observed, fusion is not likely to account for the generation of HSCs by MAPCs.


Sujet(s)
Hématopoïèse , Transplantation de cellules souches hématopoïétiques , Cellules souches multipotentes/transplantation , Animaux , Lymphocytes B/immunologie , Survie du greffon , Protéines à fluorescence verte/génétique , Hématopoïèse/immunologie , Système hématopoïétique/cytologie , Techniques in vitro , Tissu lymphoïde/cytologie , Souris , Souris de lignée C57BL , Souris de lignée NOD , Souris SCID , Souris transgéniques , Cellules souches multipotentes/immunologie , Spécificité d'organe , Protéines recombinantes/génétique , Lymphocytes T/immunologie
16.
Circulation ; 113(7): 1005-14, 2006 Feb 21.
Article de Anglais | MEDLINE | ID: mdl-16476845

RÉSUMÉ

BACKGROUND: Recent studies have shown that stem cell therapy can promote tissue regeneration; however, monitoring stem cells in vivo remains problematic owing to limitations of conventional histological assays and imaging modalities. METHODS AND RESULTS: Murine embryonic stem (ES) cells were stably transduced with a lentiviral vector carrying a novel triple-fusion (TF) reporter gene that consists of firefly luciferase, monomeric red fluorescence protein, and truncated thymidine kinase (fluc-mrfp-ttk). ES cell viability, proliferation, and differentiation ability were not adversely affected by either reporter genes or reporter probes compared with nontransduced control cells (P=NS). Afterward, 1x10(7) of ES cells carrying the TF reporter gene (ES-TF) were injected into the myocardium of adult nude rats (n=20). Control animals received nontransduced ES cells (n=6). At day 4, the bioluminescence and positron emission tomography signals in study animals were 3.7x10(7)+/-5.8x10(6) photons.s(-1).cm(-2) per steradian (sr) and 0.08+/-0.03% injected dose/g, respectively (P<0.05 versus control). Both signals increased progressively from week 1 to week 4, which indicated ES cell survival and proliferation in the host. Histological analysis demonstrated the formation of intracardiac and extracardiac teratomas. Finally, animals (n=4) that were treated with intraperitoneal injection of ganciclovir (50 mg/kg) did not develop teratomas when compared with control animals (n=4) treated with saline (1 mL/kg). CONCLUSIONS: This is the first study to characterize ES cells that stably express fluorescence, bioluminescence, and positron emission tomography reporter genes and monitor the kinetics of ES cell survival, proliferation, and migration. This versatile imaging platform should have broad applications for basic research and clinical studies on stem cell therapy.


Sujet(s)
Myocarde/cytologie , Régénération , Transplantation de cellules souches/méthodes , Cellules souches/cytologie , Animaux , Mouvement cellulaire , Prolifération cellulaire , Survie cellulaire , Embryon de mammifère/cytologie , Ganciclovir/usage thérapeutique , Gènes rapporteurs , Humains , Luminescence , Souris , Modèles animaux , Tomographie par émission de positons , Prémédication , Rats , Transplantation de cellules souches/effets indésirables , Cellules souches/physiologie , Tératome/étiologie , Tératome/prévention et contrôle , Transduction génétique , Transplantation hétérologue
17.
Proc Natl Acad Sci U S A ; 102(9): 3312-7, 2005 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-15728354

RÉSUMÉ

Leukemia inhibitory factor (LIF) is required, but not sufficient, for pluripotent mouse embryonic stem (ES) cell expansion in vitro in the absence of serum or a feeder cell layer, suggesting that additional signals are provided by serum or feeders that are necessary to support self-renewal. Here we show that transgenic ES cell lines expressing Bcl-2, an antiapoptotic protein, continue to self-renew in serum- and feeder-free conditions when supplemented with LIF; even in the absence of bone morphogenic proteins. Bcl-2-expressing clones sustain the characteristics of undifferentiated, pluripotent ES cells during long-term culture, and maintain their potential to differentiate into mature cell types. These results suggest that LIF and Bcl-2 overexpression are sufficient to expand these mouse pluripotent stem cells in vitro.


Sujet(s)
Sang , Embryon de mammifère/cytologie , Protéines proto-oncogènes c-bcl-2/métabolisme , Cellules souches/cytologie , Animaux , Apoptose/physiologie , Séquence nucléotidique , Protéines morphogénétiques osseuses/métabolisme , Division cellulaire , Chimère , Amorces ADN , Femelle , Cytométrie en flux , Humains , Immunohistochimie , Souris , Souris de lignée C57BL , Réaction de polymérisation en chaîne , Protéines proto-oncogènes c-bcl-2/physiologie
18.
Exp Hematol ; 32(11): 1051-6, 2004 Nov.
Article de Anglais | MEDLINE | ID: mdl-15539082

RÉSUMÉ

OBJECTIVE: beta1-integrins mediate hematopoietic stem and progenitor cell homing and retention in the bone marrow (BM) and inhibit hematopoietic proliferation and differentiation. Having no intrinsic kinase activity, integrins recruit intracellular kinases, such as the focal adhesion kinase (FAK) or the related proline-rich tyrosine kinase 2 (PYK2), to initiate signal transduction. Phosphatidylinositol-3-kinase (PI3K), which is involved in beta1-integrin signaling in many cell types, is physically and functionally associated with FAK in anchorage-dependent cells. Because PYK2 is the principal focal adhesion kinase expressed in primary human CD34+ cells, we assessed its functional relationship with PI3K in CD34+ cells in response to integrin engagement. METHODS: beta1-integrins on primary mobilized peripheral blood CD34+ cells and CD34+ KG1A cells were engaged by adhesion to fibronectin (FN) or by cross-linking with an anti-beta1 integrin antibody, respectively. PI3K activity and PYK2 phosphorylation were then assessed in the presence or absence of the PI3K inhibitor, wortmannin. Association between PI3K, PYK2, and the beta1-integrin subunit were also evaluated in co-immunoprecipitation experiments. RESULTS: beta1-integrin engagement induced PI3K activation, which was required for, and temporally preceded, PYK2 phosphorylation, indicating that PI3K lies upstream of PYK2 in CD34+ cells. Furthermore, although PYK2 and PI3K were constitutively associated, interaction of the PYK2/PI3K complex with beta1-integrins required prior integrin engagement and PI3K activation. CONCLUSION: Activation of PI3K following beta1-integrin engagement on human CD34+ cells results in subsequent phosphorylation of PYK2, and is required for the recruitment of the PI3K/PYK2 complex to beta1-integrins at the cell surface.


Sujet(s)
Cellules souches hématopoïétiques/métabolisme , Antigènes CD29/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protein-tyrosine kinases/métabolisme , Antigènes CD34 , Activation enzymatique/physiologie , Focal adhesion kinase 1 , Focal adhesion kinase 2 , Focal adhesion protein-tyrosine kinases , Cellules souches hématopoïétiques/enzymologie , Humains , Phosphatidylinositol 3-kinases/physiologie , Phosphorylation , Liaison aux protéines , Transport des protéines
19.
N Engl J Med ; 351(7): 657-67, 2004 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-15306667

RÉSUMÉ

BACKGROUND: The progression of chronic myelogenous leukemia (CML) to blast crisis is supported by self-renewing leukemic stem cells. In normal mouse hematopoietic stem cells, the process of self-renewal involves the beta-catenin-signaling pathway. We investigated whether leukemic stem cells in CML also use the beta-catenin pathway for self-renewal. METHODS: We used fluorescence-activated cell sorting to isolate hematopoietic stem cells, common myeloid progenitors, granulocyte-macrophage progenitors, and megakaryocyte-erythroid progenitors from marrow during several phases of CML and from normal marrow. BCR-ABL, beta-catenin, and LEF-1 transcripts were compared by means of a quantitative reverse-transcriptase-polymerase-chain-reaction assay in normal and CML hematopoietic stem cells and granulocyte-macrophage progenitors. Confocal fluorescence microscopy and a lymphoid enhancer factor/T-cell factor reporter assay were used to detect nuclear beta-catenin in these cells. In vitro replating assays were used to identify self-renewing cells as candidate leukemic stem cells, and the dependence of self-renewal on beta-catenin activation was tested by lentiviral transduction of hematopoietic progenitors with axin, an inhibitor of the beta-catenin pathway. RESULTS: The granulocyte-macrophage progenitor pool from patients with CML in blast crisis and imatinib-resistant CML was expanded, expressed BCR-ABL, and had elevated levels of nuclear beta-catenin as compared with the levels in progenitors from normal marrow. Unlike normal granulocyte-macrophage progenitors, CML granulocyte-macrophage progenitors formed self-renewing, replatable myeloid colonies, and in vitro self-renewal capacity was reduced by enforced expression of axin. CONCLUSIONS: Activation of beta-catenin in CML granulocyte-macrophage progenitors appears to enhance the self-renewal activity and leukemic potential of these cells.


Sujet(s)
Crise blastique/physiopathologie , Protéines du cytosquelette/métabolisme , Cellules souches hématopoïétiques/physiologie , Leucémie myéloïde chronique BCR-ABL positive/physiopathologie , Transactivateurs/métabolisme , Adulte , Sujet âgé , Antinéoplasiques/usage thérapeutique , Benzamides , Test clonogénique , Protéines de liaison à l'ADN/métabolisme , Résistance aux médicaments antinéoplasiques , Femelle , Protéines de fusion bcr-abl/métabolisme , Granulocytes/cytologie , Cellules souches hématopoïétiques/métabolisme , Humains , Mésilate d'imatinib , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Facteur de transcription LEF-1 , Macrophages/cytologie , Mâle , Microscopie confocale , Adulte d'âge moyen , Pipérazines/usage thérapeutique , Pyrimidines/usage thérapeutique , ARN tumoral , RT-PCR , Facteurs de transcription/métabolisme , bêta-Caténine
20.
Exp Hematol ; 32(4): 365-74, 2004 Apr.
Article de Anglais | MEDLINE | ID: mdl-15050747

RÉSUMÉ

OBJECTIVE: Hematopoietic progenitor proliferation and differentiation are inhibited by integrin engagement of fibronectin (FN). Focal adhesion kinases have been shown to mediate intracellular signaling from integrins, and we recently demonstrated that gene expression and pre-mRNA splicing of the focal adhesion kinase, PYK2, is abnormal in CD34(+) cells from chronic myelogenous leukemia (CML) patients. Here we investigated whether PYK2 gene products mediate integrin signaling in hematopoietic stem and progenitor cells. METHODS: Cord blood CD34(+) cells were retrovirally transduced with vectors encoding Pyk2H, Pyk2, or the dominant negative-acting, kinase-deficient, C-terminal PYK2 fragment, PRNK, and myeloid proliferation and differentiation was assessed using colony-forming cell (CFC), long-term culture-initiating cell (LTC-IC), and liquid culture assays. RESULTS: CD34(+) cells overexpressing Pyk2H or Pyk2 generated 50% less colony-forming unit granulocyte/macrophage (CFU-GM) than eGFP-transduced controls. Although the number of CFC generated by PRNK-expressing cells was unchanged, LTC-IC were significantly reduced. Culture of CD34(+) cells on FN significantly reduced the generation of mature myeloid cells vs those cultured on BSA-coated wells, and could be overcome by addition of SCF. As is observed when integrins are engaged, overexpression of either Pyk2H or Pyk2 decreased committed myeloid progenitor proliferation and differentiation; however, SCF could not override this inhibition. Finally, as is observed when integrins are not engaged, PRNK-mediated inhibition of endogenous Pyk2H resulted in integrin-nonresponsive proliferation and differentiation of myeloid precursors and accelerated differentiation of primitive hematopoietic progenitors. CONCLUSION: These studies indicate that PYK2 gene products mediate integrin-induced signals that regulate myelopoiesis.


Sujet(s)
Intégrines/physiologie , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Myélopoïèse/physiologie , Protéines tumorales/physiologie , Protein-tyrosine kinases/physiologie , Animaux , Bovins , Différenciation cellulaire/effets des médicaments et des substances chimiques , Division cellulaire/effets des médicaments et des substances chimiques , Test clonogénique , Milieux de culture , Fibronectines , Focal adhesion kinase 2 , Cellules HL-60 , Humains , Isoenzymes/génétique , Isoenzymes/physiologie , Myélopoïèse/effets des médicaments et des substances chimiques , Fragments peptidiques/pharmacologie , Phosphorylation , Maturation post-traductionnelle des protéines , Protein-tyrosine kinases/génétique , Protéines de fusion recombinantes/physiologie , Sérumalbumine bovine , Transduction du signal , Facteur de croissance des cellules souches/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...