Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cells ; 10(10)2021 09 23.
Article in English | MEDLINE | ID: mdl-34685497

ABSTRACT

Autophagy has been described as harboring a dual role in cancer development and therapy. Depending on the context, it can exert either pro-survival or pro-death functions. Here, we review what is known about autophagy in crizotinib-treated ALK+ ALCL. We first present our main findings on the role and regulation of autophagy in these cells. Then, we provide literature-driven hypotheses that could explain mechanistically the pro-survival properties of autophagy in crizotinib-treated bulk and stem-like ALK+ ALCL cells. Finally, we discuss how the potentiation of autophagy, which occurs with combined therapies (ALK and BCL2 or ALK and RAF1 co-inhibition), could convert it from a survival mechanism to a pro-death process.


Subject(s)
Autophagy/drug effects , Crizotinib/pharmacology , Drug Resistance, Neoplasm/drug effects , Lymphoma/drug therapy , Anaplastic Lymphoma Kinase/drug effects , Humans , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/drug effects
2.
Cancers (Basel) ; 13(2)2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33430343

ABSTRACT

Previously it was shown that autophagy contributes to crizotinib resistance in ALK-positive anaplastic large cell lymphoma (ALK + ALCL). We asked if autophagy is equally important in two distinct subsets of ALK + ALCL, namely Reporter Unresponsive (RU) and Reporter Responsive (RR), of which RR cells display stem-like properties. Autophagic flux was assessed with a fluorescence tagged LC3 reporter and immunoblots to detect endogenous LC3 alongside chloroquine, an autophagy inhibitor. The stem-like RR cells displayed significantly higher autophagic response upon crizotinib treatment. Their exaggerated autophagic response is cytoprotective against crizotinib, as inhibition of autophagy using chloroquine or shRNA against BECN1 or ATG7 led to a decrease in their viability. In contrast, autophagy inhibition in RU resulted in minimal changes. Since the differential protein expression of MYC is a regulator of the RU/RR dichotomy and is higher in RR cells, we asked if MYC regulates the autophagy-mediated cytoprotective effect. Inhibition of MYC in RR cells using shRNA significantly blunted crizotinib-induced autophagic response and effectively suppressed this cytoprotective effect. In conclusion, stem-like RR cells respond with rapid and intense autophagic flux which manifests with crizotinib resistance. For the first time, we have highlighted the direct role of MYC in regulating autophagy and its associated chemoresistance phenotype in ALK + ALCL stem-like cells.

3.
Cancers (Basel) ; 12(10)2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33066037

ABSTRACT

Anaplastic lymphoma kinase positive anaplastic large cell lymphomas (ALK+ ALCL) are an aggressive pediatric disease. The therapeutic options comprise chemotherapy, which is efficient in approximately 70% of patients, and targeted therapies, such as crizotinib (an ALK tyrosine kinase inhibitor (TKI)), used in refractory/relapsed cases. Research efforts have also converged toward the development of combined therapies to improve treatment. In this context, we studied whether autophagy could be modulated to improve crizotinib therapy. Autophagy is a vesicular recycling pathway, known to be associated with either cell survival or cell death depending on the cancer and therapy. We previously demonstrated that crizotinib induced cytoprotective autophagy in ALK+ lymphoma cells and that its further intensification was associated with cell death. In line with these results, we show here that combined ALK and Rapidly Accelerated Fibrosarcoma 1 (RAF1) inhibition, using pharmacological (vemurafenib) or molecular (small interfering RNA targeting RAF1 (siRAF1) or microRNA-7-5p (miR-7-5p) mimics) strategies, also triggered autophagy and potentiated the toxicity of TKI. Mechanistically, we found that this combined therapy resulted in the decrease of the inhibitory phosphorylation on Unc-51-like kinase-1 (ULK1) (a key protein in autophagy initiation), which may account for the enforced autophagy and cytokilling effect. Altogether, our results support the development of ALK and RAF1 combined inhibition as a new therapeutic approach in ALK+ ALCL.

4.
Cells ; 8(2)2019 01 30.
Article in English | MEDLINE | ID: mdl-30704144

ABSTRACT

Haematopoiesis is a tightly orchestrated process where a pool of hematopoietic stem and progenitor cells (HSPCs) with high self-renewal potential can give rise to both lymphoid and myeloid lineages. The HSPCs pool is reduced with ageing resulting in few HSPC clones maintaining haematopoiesis thereby reducing blood cell diversity, a phenomenon called clonal haematopoiesis. Clonal expansion of HSPCs carrying specific genetic mutations leads to increased risk for haematological malignancies. Therefore, it comes as no surprise that hematopoietic tumours develop in higher frequency in elderly people. Unfortunately, elderly patients with leukaemia or lymphoma still have an unsatisfactory prognosis compared to younger ones highlighting the need to develop more efficient therapies for this group of patients. Growing evidence indicates that macroautophagy (hereafter referred to as autophagy) is essential for health and longevity. This review is focusing on the role of autophagy in normal haematopoiesis as well as in leukaemia and lymphoma development. Attenuated autophagy may support early hematopoietic neoplasia whereas activation of autophagy in later stages of tumour development and in response to a variety of therapies rather triggers a pro-tumoral response. Novel insights into the role of autophagy in haematopoiesis will be discussed in light of designing new autophagy modulating therapies in hematopoietic cancers.


Subject(s)
Autophagy , Leukemia/pathology , Lymphoma/pathology , Animals , Clinical Trials as Topic , Hematopoiesis , Humans
5.
Haematologica ; 104(7): 1428-1439, 2019 07.
Article in English | MEDLINE | ID: mdl-30679328

ABSTRACT

Anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphomas are tumors that carry translocations involving the ALK gene at the 2p23 locus, leading to the expression of ALK tyrosine kinase fusion oncoproteins. Amongst hematologic malignancies, these lymphomas are particular in that they express very low levels of B-cell lymphoma 2 (BCL2), a recognized inhibitor of apoptosis and autophagy, two processes that share complex interconnections. We have previously shown that treatment of ALK-positive anaplastic large cell lymphoma cells with the ALK tyrosine kinase inhibitor crizotinib induces autophagy as a pro-survival response. Here, we observed that crizotinib-mediated inactivation of ALK caused an increase in BCL2 levels that restrained the cytotoxic effects of the drug. BCL2 downregulation in combination with crizotinib treatment potentiated loss of cell viability through both an increase in autophagic flux and cell death, including apoptosis. More importantly, our data revealed that the blockade of autophagic flux completely reversed impaired cell viability, which demonstrates that excessive autophagy is associated with cell death. We propose that the downregulation of BCL2 protein, which plays a central role in the autophagic and apoptotic machinery, combined with crizotinib treatment may represent a promising therapeutic alternative to current ALK-positive anaplastic large cell lymphoma treatments.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Antineoplastic Agents/pharmacology , Autophagy , Crizotinib/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lymphoma, Large-Cell, Anaplastic/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Anaplastic Lymphoma Kinase/genetics , Animals , Cell Death , Cell Proliferation/drug effects , Humans , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Prognosis , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cancers (Basel) ; 9(12)2017 Nov 27.
Article in English | MEDLINE | ID: mdl-29186933

ABSTRACT

Autophagy is an evolutionarily conserved catabolic process, which is used by the cells for cytoplasmic quality control. This process is induced following different kinds of stresses e.g., metabolic, environmental, or therapeutic, and acts, in this framework, as a cell survival mechanism. However, under certain circumstances, autophagy has been associated with cell death. This duality has been extensively reported in solid and hematological cancers, and has been observed during both tumor development and cancer therapy. As autophagy plays a critical role at the crossroads between cell survival and cell death, its involvement and therapeutic modulation (either activation or inhibition) are currently intensively studied in cancer biology, to improve treatments and patient outcomes. Over the last few years, studies have demonstrated the occurrence of autophagy in different Anaplastic Lymphoma Kinase (ALK)-associated cancers, notably ALK-positive anaplastic large cell lymphoma (ALCL), non-small cell lung carcinoma (NSCLC), Neuroblastoma (NB), and Rhabdomyosarcoma (RMS). In this review, we will first briefly describe the autophagic process and how it can lead to opposite outcomes in anti-cancer therapies, and we will then focus on what is currently known regarding autophagy in ALK-associated cancers.

7.
Med Sci (Paris) ; 33(3): 328-334, 2017 Mar.
Article in French | MEDLINE | ID: mdl-28367821

ABSTRACT

Autophagy is a self-cannibalism process essential for tissue homeostasis, which can be activated following different environmental stressful conditions. In normal cells, autophagy could act as a brake to prevent tumorigenesis, but cancer cells are able to hijack this process to their own benefit, to promote tumor growth and/or tumor resistance to anti-cancer therapies. Scientists and clinicians attempt to modulate this process to improve therapies, using autophagy inhibitors or activators, some of them being tested currently in clinical trials against several types of tumors. Thus, it appears that autophagy is at the center of a showdown between cancer cells and anti-cancer therapies. In this review, we focus on the mechanisms by which autophagy could be either the yin or the yang of cancers.


Subject(s)
Autophagy/physiology , Cell Transformation, Neoplastic , Neoplasms/pathology , Animals , Cell Survival , Cell Transformation, Neoplastic/pathology , Humans
8.
Oncotarget ; 6(30): 30149-64, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26338968

ABSTRACT

Anaplastic Lymphoma Kinase-positive Anaplastic Large Cell Lymphomas (ALK+ ALCL) occur predominantly in children and young adults. Their treatment, based on aggressive chemotherapy, is not optimal since ALCL patients can still expect a 30% 2-year relapse rate. Tumor relapses are very aggressive and their underlying mechanisms are unknown. Crizotinib is the most advanced ALK tyrosine kinase inhibitor and is already used in clinics to treat ALK-associated cancers. However, crizotinib escape mechanisms have emerged, thus preventing its use in frontline ALCL therapy. The process of autophagy has been proposed as the next target for elimination of the resistance to tyrosine kinase inhibitors. In this study, we investigated whether autophagy is activated in ALCL cells submitted to ALK inactivation (using crizotinib or ALK-targeting siRNA). Classical autophagy read-outs such as autophagosome visualization/quantification by electron microscopy and LC3-B marker turn-over assays were used to demonstrate autophagy induction and flux activation upon ALK inactivation. This was demonstrated to have a cytoprotective role on cell viability and clonogenic assays following combined ALK and autophagy inhibition. Altogether, our results suggest that co-treatment with crizotinib and chloroquine (two drugs already used in clinics) could be beneficial for ALK-positive ALCL patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Chloroquine/pharmacology , Lymphoma, Large-Cell, Anaplastic/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Animals , Cell Line, Tumor , Cell Survival/drug effects , Crizotinib , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Lymphoma, Large-Cell, Anaplastic/enzymology , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/pathology , Mice, Inbred NOD , Mice, SCID , Microtubule-Associated Proteins/metabolism , RNA Interference , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Xenograft Model Antitumor Assays
9.
J Clin Invest ; 125(9): 3505-18, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26258416

ABSTRACT

The regulatory microRNA miR-150 is involved in the development of hemopathies and is downregulated in T-lymphomas, such as anaplastic large-cell lymphoma (ALCL) tumors. ALCL is defined by the presence or absence of translocations that activate the anaplastic lymphoma kinase (ALK), with nucleophosmin-ALK (NPM-ALK) fusions being the most common. Here, we compared samples of primary NPM-ALK(+) and NPM-ALK(-) ALCL to investigate the role of miR-150 downstream of NPM-ALK. Methylation of the MIR150 gene was substantially elevated in NPM-ALK(+) biopsies and correlated with reduced miR-150 expression. In NPM-ALK(+) cell lines, DNA hypermethylation-mediated miR-150 repression required ALK-dependent pathways, as ALK inhibition restored miR-150 expression. Moreover, epigenetic silencing of miR-150 was due to the activation of STAT3, a major downstream substrate of NPM-ALK, in cooperation with DNA methyltransferase 1 (DNMT1). Accordingly, miR-150 repression was turned off following treatment with the DNMT inhibitor, decitabine. In murine NPM-ALK(+) xenograft models, miR-150 upregulation induced antineoplastic activity. Treatment of crizotinib-resistant NPM-ALK(+) KARPAS-299-CR06 cells with decitabine or ectopic miR-150 expression reduced viability and growth. Altogether, our results suggest that hypomethylating drugs, alone or in combination with other agents, may benefit ALK(+) patients harboring tumors resistant to crizotinib and other anti-ALK tyrosine kinase inhibitors (TKIs). Moreover, these results support further work on miR-150 in these and other ALK(+) malignancies.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Gene Silencing , Lymphoma, Large-Cell, Anaplastic/metabolism , MicroRNAs/biosynthesis , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , RNA, Neoplasm/biosynthesis , Animals , Cell Line, Tumor , Crizotinib , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Humans , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/pathology , Male , Mice , Mice, Transgenic , MicroRNAs/genetics , Protein-Tyrosine Kinases/genetics , RNA, Neoplasm/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
10.
Front Biosci (Schol Ed) ; 7(2): 236-47, 2015 06 01.
Article in English | MEDLINE | ID: mdl-25961699

ABSTRACT

Our current understanding of oncogenic Anaplastic Lymphoma Kinase (ALK)-induced lymphomagenesis has relied for over 20 years on multiple and complementary studies performed on various experimental models, encompassing ALK oncogene expressing cells, their grafts into immune-compromised mice, the generation of genetically engineered mouse models (GEMMs) and, when available, the use of patient samples from Anaplastic Large Cell Lymphoma (ALCL) tumour banks. Of note, and to our knowledge, no ALK-positive ALCL 3D culture system has been described so far. In this review, we will first outline how these different cell and mouse models were designed, and what key findings they revealed (or confirmed) towards oncogenic ALK-induced lymphomagenesis. Secondly, we will discuss how recent and revolutionary advances in genetic engineering technology are likely to complete our understanding of ALK-related disease in an effort to improve current therapeutic approaches.


Subject(s)
Disease Models, Animal , Lymphoma, Large-Cell, Anaplastic/enzymology , Lymphoma, Large-Cell, Anaplastic/genetics , Nuclear Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Animals , Humans , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nucleophosmin , Protein-Tyrosine Kinases , Receptor Protein-Tyrosine Kinases/genetics
11.
Blood ; 119(20): 4698-707, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22394598

ABSTRACT

Anaplastic large-cell lymphomas (ALCLs) bearing the t(2;5) translocation (ALK(+)ALCLs) are frequently characterized by skin colonization and associated with a poor prognosis. Using conditional transgenic models of anaplastic lymphoma kinase-positive (ALK(+)) lymphomas and human ALK(+)ALCL cell lines, in the present study, we show that high-mobility-group box-1 (HMGB-1), a proinflammatory cytokine, is released by ALK(+) cells, and demonstrate extracellular HMGB-1-stimulated secretion of the IL-8 chemokine by HaCaT keratinocytes through the involvement of MMP-9, PAR-2, and the NF-κB pathway. Furthermore, we demonstrate that, in vitro, IL-8 is able to induce the invasiveness of ALK(+) cells, which express the IL-8 receptors CXCR1 and CXCR2. In vitro and in vivo, HMGB-1 inhibition achieved by glycyrrhizin treatment led to a drastic reduction in ALK(+) cell invasiveness. The pathophysiological relevance of our observations was confirmed by demonstrating that the HMGB-1 and IL-8 receptors are expressed in ALK(+)ALCL biopsies. We have also shown that IL-8 secretion is correlated with leukemic dissemination of ALK(+) cells in a significant number of patients. The results of the present study demonstrate for the first time a relationship among the pro-inflammatory mediators HMGB-1, MMP-9, PAR-2, and IL-8. We propose that these mediators create a premetastatic niche within the skin, thereby participating in ALK(+) lymphoma epidermotropism.


Subject(s)
HMGB1 Protein/physiology , Interleukin-8/metabolism , Keratinocytes/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , NF-kappa B/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Skin/metabolism , Anaplastic Lymphoma Kinase , Animals , Cells, Cultured , Female , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Keratinocytes/pathology , Leukemic Infiltration/genetics , Leukemic Infiltration/metabolism , Leukemic Infiltration/pathology , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/physiology , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , NF-kappa B/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , Receptor, PAR-2/physiology , Signal Transduction/physiology , Skin/pathology , Stem Cell Niche/genetics , Stem Cell Niche/immunology
12.
J Exp Med ; 208(5): 973-86, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21482699

ABSTRACT

Angiogenesis plays an essential role in several diseases of the eye and in the growth of solid tumors, but existing antiangiogenic therapies have limited benefits in several cases. We report the antiangiogenic effects of a monoclonal antibody, CL1-R2, in several animal models of neovascularization. CL1-R2 recognizes human CD160, a membrane receptor which is conserved in various mammal species. We show that CD160 is expressed on the endothelial cells of newly formed blood vessels in human colon carcinoma and mouse B16 melanoma but not in vessels of healthy tissues. CL1-R2 reduced fibroblast growth factor 2-induced neovascularization in the rabbit cornea, in a mouse model of oxygen-induced retinopathy, and in a mouse Matrigel plug assay. Treatment of B16 melanoma-bearing mice with CL1-R2 combined with cyclophosphamide chemotherapy caused regression of the tumor vasculature and normalization of the remaining vessels as shown by Doppler ultrasonography, intravital microscopy, and histology. These studies validate CD160 as a potential new target in cases of human pathological ocular and tumor neoangiogenesis that do not respond or become resistant to existing antiangiogenic drugs.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD/metabolism , Neovascularization, Pathologic/drug therapy , Receptors, Immunologic/metabolism , Animals , Antigens, CD/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Corneal Neovascularization/drug therapy , Corneal Neovascularization/genetics , Corneal Neovascularization/metabolism , Corneal Neovascularization/pathology , Cyclophosphamide/pharmacology , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Male , Melanoma/blood supply , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Rabbits , Receptors, Immunologic/genetics , Retinal Diseases/drug therapy , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Diseases/pathology
13.
Blood ; 115(20): 4061-70, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20223922

ABSTRACT

NPM-ALK (nucleophosmin-anaplastic lymphoma kinase) and TPM3-ALK (nonmuscular tropomyosin 3-anaplastic lymphoma kinase) are oncogenic tyrosine kinases implicated in the pathogenesis of human ALK-positive lymphoma. We report here the development of novel conditional mouse models for ALK-induced lymphomagenesis, with the use of the tetracycline regulatory system under the control of the EmuSRalpha enhancer/promoter. The expression of either oncogene resulted in the arrest of the differentiation of early B cells and lymphomagenesis. We also observed the development of skin keratoacanthoma lesions, probably because of aberrant ALK expression in keratinocytes. The inactivation of the ALK oncogene on doxycycline treatment was sufficient to induce sustained regression of both hematopoietic tumors and skin disease. Importantly, treatment with the specific ALK inhibitor (PF-2341066) also reversed the pathologic states, showing the value of these mouse models for the validation of ALK tyrosine kinase inhibitors. Thus, our results show (1) that NPM-ALK and TPM3-ALK oncogenes are sufficient for lymphoma/leukemia development and required for tumor maintenance, hence validating ALK as potentially effective therapeutic target; and (2) for the first time, in vivo, the equal tumorigenic potential of the NPM-ALK and TPM3-ALK oncogenic tyrosine kinases. Our models offer a new tool to investigate in vivo the molecular mechanisms associated with ALK-induced lymphoproliferative disorders.


Subject(s)
Leukemia, B-Cell/pathology , Lymphoma, B-Cell/pathology , Protein-Tyrosine Kinases/genetics , Tropomyosin/genetics , Anaplastic Lymphoma Kinase , Animals , Apoptosis , Blotting, Western , Cell Differentiation , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoblotting , Immunoenzyme Techniques , Integrases/metabolism , Leukemia, B-Cell/genetics , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Mice , Mice, Transgenic , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases , Reverse Transcriptase Polymerase Chain Reaction , Tropomyosin/metabolism
14.
PLoS One ; 3(5): e2125, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18461184

ABSTRACT

BACKGROUND: Conditional transgenic models have established that tumors require sustained oncogene activation for tumor maintenance, exhibiting the phenomenon known as "oncogene-addiction." However, most cancers are caused by multiple genetic events making it difficult to determine which oncogenes or combination of oncogenes will be the most effective targets for their treatment. METHODOLOGY/PRINCIPAL FINDINGS: To examine how the MYC and K-ras(G12D) oncogenes cooperate for the initiation and maintenance of tumorigenesis, we generated double conditional transgenic tumor models of lung adenocarcinoma and lymphoma. The ability of MYC and K-ras(G12D) to cooperate for tumorigenesis and the ability of the inactivation of these oncogenes to result in tumor regression depended upon the specific tissue context. MYC-, K-ras(G12D)- or MYC/K-ras(G12D)-induced lymphomas exhibited sustained regression upon the inactivation of either or both oncogenes. However, in marked contrast, MYC-induced lung tumors failed to regress completely upon oncogene inactivation; whereas K-ras(G12D)-induced lung tumors regressed completely. Importantly, the combined inactivation of both MYC and K-ras(G12D) resulted more frequently in complete lung tumor regression. To account for the different roles of MYC and K-ras(G12D) in maintenance of lung tumors, we found that the down-stream mediators of K-ras(G12D) signaling, Stat3 and Stat5, are dephosphorylated following conditional K-ras(G12D) but not MYC inactivation. In contrast, Stat3 becomes dephosphorylated in lymphoma cells upon inactivation of MYC and/or K-ras(G12D). Interestingly, MYC-induced lung tumors that failed to regress upon MYC inactivation were found to have persistent Stat3 and Stat5 phosphorylation. CONCLUSIONS/SIGNIFICANCE: Taken together, our findings point to the importance of the K-Ras and associated down-stream Stat effector pathways in the initiation and maintenance of lymphomas and lung tumors. We suggest that combined targeting of oncogenic pathways is more likely to be effective in the treatment of lung cancers and lymphomas.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing/physiology , Genes, myc , Genes, ras , Lung Neoplasms/genetics , Lymphoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Doxycycline/therapeutic use , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphoma/immunology , Mice , Mice, Transgenic , Respiratory Mucosa/drug effects
15.
Cancer Biol Ther ; 6(8): 1318-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17660712

ABSTRACT

Overexpression and activation of TPM3-ALK tyrosine kinase fusion protein is a causal oncogenic event in the development of Anaplastic Large Cell Lymphoma and Inflammatory Myofibroblastic ALK-positive tumors. Thus, the development of ALK specific tyrosine kinase inhibitors is a current therapeutic challenge. Animal models are essential to assess, in vivo, the efficiency of ALK-oncogene inhibitors and to identify new and/or additional therapeutic targets in the ALK tumorigenesis pathway. Using the tetracycline system to allow conditional and concomitant TPM3-ALK and luciferase expression, we have developed a unique transplant model for bioluminescent TPM3-ALK-induced fibroblastic tumors in athymic nude mice. The reversible TPM3-ALK expression allowed us to demonstrate that this oncogene is essential for the tumor growth and its maintenance. In addition, we showed that this model could be used to precisely assess tumor growth inhibition upon ALK chemical inactivation. As proof of principle, we used the general tyrosine kinase inhibitor herbimycin A to inhibit ALK oncoprotein activity. As expected, herbimycin A treatment reduced tumor growth as assessed both by tumor volume measurement and bioluminescent imaging. We conclude that this transplant model for TPM3-ALK-induced tumors represents a valuable tool not only to accurately and rapidly evaluate in vivo ALK-targeted therapies but also to gain insight into the mechanism of ALK-positive tumor development.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Disease Models, Animal , Lymphoma, Large-Cell, Anaplastic/drug therapy , Mice , Oncogene Proteins, Fusion/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Tropomyosin/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Animals , Antibiotics, Antineoplastic/therapeutic use , Benzoquinones/pharmacology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Drug Screening Assays, Antitumor/methods , Genes, Reporter , Lactams, Macrocyclic/pharmacology , Luciferases/analysis , Luciferases/genetics , Luminescent Agents/analysis , Lymphoma, Large-Cell, Anaplastic/enzymology , Mice, Nude , Neoplasm Transplantation , Oncogene Proteins, Fusion/analysis , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases , Rifabutin/analogs & derivatives , Tropomyosin/analysis
16.
Blood ; 109(5): 2156-64, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17077326

ABSTRACT

With the use of microarray gene-expression profiling, we analyzed a homogeneous series of 32 patients with systemic anaplastic large-cell lymphoma (ALCL) and 5 ALCL cell lines. Unsupervised analysis classified ALCL in 2 clusters, corresponding essentially to morphologic subgroups (ie, common type vs small cell and "mixed" variants) and clinical variables. Patients with a morphologic variant of ALCL had advanced-stage disease. This group included a significant number of patients who experienced early relapse. Supervised analysis showed that ALK+ALCL and ALK- ALCL have different gene-expression profiles, further confirming that they are different entities. Among the most significantly differentially expressed genes between ALK+ and ALK- samples, we found BCL6, PTPN12, CEBPB, and SERPINA1 genes to be overexpressed in ALK+ ALCL. This result was confirmed at the protein level for BCL-6, C/EBPbeta and serpinA1 through tissue microarrays. The molecular signature of ALK- ALCL included overexpression of CCR7, CNTFR, IL22, and IL21 genes but did not provide any obvious clues to the molecular mechanism underlying this tumor subtype. Once confirmed on a larger number of patients, the results of the present study could be used for clinical and therapeutic management of patients at the time of diagnosis.


Subject(s)
Cell Shape , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/pathology , Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Humans , Immunohistochemistry , Lymphoma, Large-Cell, Anaplastic/classification , Lymphoma, Large-Cell, Anaplastic/enzymology , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases , Tissue Array Analysis
17.
Proc Natl Acad Sci U S A ; 103(44): 16266-71, 2006 Oct 31.
Article in English | MEDLINE | ID: mdl-17056717

ABSTRACT

The targeted inactivation of oncogenes offers a rational therapeutic approach for the treatment of cancer. However, the therapeutic inactivation of a single oncogene has been associated with tumor recurrence. Therefore, it is necessary to develop strategies to override mechanisms of tumor escape from oncogene dependence. We report here that the targeted inactivation of MYC is sufficient to induce sustained regression of hematopoietic tumors in transgenic mice, except in tumors that had lost p53 function. p53 negative tumors were unable to be completely eliminated, as demonstrated by the kinetics of tumor cell elimination revealed by bioluminescence imaging. Histological examination revealed that upon MYC inactivation, the loss of p53 led to a deficiency in thrombospondin-1 (TSP-1) expression, a potent antiangiogenic protein, and the subsequent inability to shut off angiogenesis. Restoration of p53 expression in these tumors re-established TSP-1 expression. This permitted the suppression of angiogenesis and subsequent sustained tumor regression upon MYC inactivation. Similarly, the restoration of TSP-1 alone in p53 negative tumors resulted in the shut down of angiogenesis and led to sustained tumor regression upon MYC inactivation. Hence, the complete regression of tumor mass driven by inactivation of the MYC oncogene requires the p53-dependent induction of TSP-1 and the shut down of angiogenesis. Notably, overexpression of TSP-1 alone did not influence tumor growth. Therefore, the combined inactivation of oncogenes and angiogenesis may be a more clinically effective treatment of cancer. We conclude that angiogenesis is an essential component of oncogene addiction.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Thrombospondin 1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Mice , Neoplasms/blood supply , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
18.
J Exp Med ; 202(11): 1599-611, 2005 Dec 05.
Article in English | MEDLINE | ID: mdl-16330818

ABSTRACT

Knowledge of the molecular networks controlling the proliferation and fate of hematopoietic stem cells (HSC) is essential to understand their function in maintaining blood cell production during normal hematopoiesis and upon clinical transplantation. Using highly purified stem and progenitor cell populations, we define the proliferation index and status of the cell cycle machinery at discrete stages of hematopoietic differentiation and during cytokine-mediated HSC mobilization. We identify distinct sets of cell cycle proteins that specifically associate with differentiation, self-renewal, and maintenance of quiescence in HSC and progenitor cells. Moreover, we describe a striking inequality of function among in vivo cycling and quiescent HSC by demonstrating that their long-term engraftment potential resides predominantly in the G(0) fraction. These data provide a direct link between HSC proliferation and function and identify discrete molecular targets in regulating HSC cell fate decisions that could have implications for both the therapeutic use of HSC and the understanding of leukemic transformation.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Resting Phase, Cell Cycle/physiology , Animals , Cell Separation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Profiling , Graft Survival/physiology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Leukemia/metabolism , Leukemia/pathology , Leukemia/therapy , Mice , Reverse Transcriptase Polymerase Chain Reaction
19.
Oncogene ; 24(40): 6101-7, 2005 Sep 08.
Article in English | MEDLINE | ID: mdl-16007205

ABSTRACT

Microarray-based formats offer a high-resolution alternative to conventional, chromosome-based comparative genomic hybridization (CGH) methods for assessing DNA copy number alteration (CNA) genome-wide in human cancer. For murine tumors, array CGH should provide even greater advantage, since murine chromosomes are more difficult to individually discern. We report here the adaptation and evaluation of a cDNA microarray-based CGH method for the routine characterization of CNAs in murine tumors, using mouse cDNA microarrays representing approximately 14,000 different genes, thereby providing an average mapping resolution of 109 kb. As a first application, we have characterized CNAs in a set of 10 primary and recurrent lymphomas derived from a Myc-induced murine lymphoma model. In primary lymphomas and more commonly in Myc-independent relapses, we identified a recurrent genomic DNA loss at chromosome 3G3-3H4, and recurrent amplifications at chromosome 3F2.1-3G3 and chromosome 15E1/E2-15F3, the boundaries of which we defined with high resolution. Further, by profiling gene expression using the same microarray platform, we identified within CNAs the relevant subset of candidate cancer genes displaying comparably altered expression, including Mcl1 (myeloid cell leukemia sequence 1), a highly expressed antiapoptotic gene residing within the chr 3 amplicon peak. CGH on mouse cDNA microarrays therefore represents a reliable method for the high-resolution characterization of CNAs in murine tumors, and a powerful approach for elucidating the molecular events in tumor development and progression in murine models.


Subject(s)
Chromosome Mapping , Lymphoma/genetics , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis/methods , Animals , Disease Models, Animal , Disease Progression , Female , Gene Amplification , Gene Deletion , Genes, myc , Lymphoma/pathology , Male , Mice , Mice, Inbred C57BL , Reproducibility of Results
20.
Semin Cancer Biol ; 14(1): 3-11, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14757531

ABSTRACT

The ability to model cancer in the mouse has provided a robust methodology to dissect the molecular etiology of cancer. These models serve as potentially powerful platforms to preclinically evaluate novel therapeutics. In particular, the recent development of strategies to conditionally induce the or knockout the function of genes in a tissue specific manner has enabled investigators to engineer mice to demonstrate that the targeted inactivation of specific oncogenes can be effective in inducing sustained regression of tumors. Thus, these animal models will be useful to define the specific genes that will be therapeutically useful to target for the treatment of particular human cancers.


Subject(s)
Models, Animal , Neoplasms/genetics , Neoplasms/therapy , Oncogenes/genetics , Animals , Gene Expression Regulation , Genetic Therapy/methods , Humans , Neoplasms/metabolism , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...