Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Cancer Cell ; 41(11): 1892-1910.e10, 2023 11 13.
Article En | MEDLINE | ID: mdl-37863068

Liver metastases are associated with poor response to current pharmacological treatments, including immunotherapy. We describe a lentiviral vector (LV) platform to selectively engineer liver macrophages, including Kupffer cells and tumor-associated macrophages (TAMs), to deliver type I interferon (IFNα) to liver metastases. Gene-based IFNα delivery delays the growth of colorectal and pancreatic ductal adenocarcinoma liver metastases in mice. Response to IFNα is associated with TAM immune activation, enhanced MHC-II-restricted antigen presentation and reduced exhaustion of CD8+ T cells. Conversely, increased IL-10 signaling, expansion of Eomes CD4+ T cells, a cell type displaying features of type I regulatory T (Tr1) cells, and CTLA-4 expression are associated with resistance to therapy. Targeting regulatory T cell functions by combinatorial CTLA-4 immune checkpoint blockade and IFNα LV delivery expands tumor-reactive T cells, attaining complete response in most mice. These findings support a promising therapeutic strategy with feasible translation to patients with unmet medical need.


CD8-Positive T-Lymphocytes , Liver Neoplasms , Humans , Mice , Animals , CTLA-4 Antigen/metabolism , Tumor Microenvironment/genetics , Macrophages , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Liver Neoplasms/pathology
2.
EMBO Mol Med ; 15(11): e17810, 2023 11 08.
Article En | MEDLINE | ID: mdl-37807875

One of the defining features of acute myeloid leukemia (AML) is an arrest of myeloid differentiation whose molecular determinants are still poorly defined. Pharmacological removal of the differentiation block contributes to the cure of acute promyelocytic leukemia (APL) in the absence of cytotoxic chemotherapy, but this approach has not yet been translated to non-APL AMLs. Here, by investigating the function of hypoxia-inducible transcription factors HIF1α and HIF2α, we found that both genes exert oncogenic functions in AML and that HIF2α is a novel regulator of the AML differentiation block. Mechanistically, we found that HIF2α promotes the expression of transcriptional repressors that have been implicated in suppressing AML myeloid differentiation programs. Importantly, we positioned HIF2α under direct transcriptional control by the prodifferentiation agent all-trans retinoic acid (ATRA) and demonstrated that HIF2α blockade cooperates with ATRA to trigger AML cell differentiation. In conclusion, we propose that HIF2α inhibition may open new therapeutic avenues for AML treatment by licensing blasts maturation and leukemia debulking.


Leukemia, Myeloid, Acute , Leukemia, Promyelocytic, Acute , Humans , Transcription Factors/metabolism , Leukemia, Myeloid, Acute/drug therapy , Tretinoin/pharmacology , Tretinoin/metabolism , Tretinoin/therapeutic use , Gene Expression Regulation , Cell Differentiation , Leukemia, Promyelocytic, Acute/drug therapy
3.
Trends Mol Med ; 29(4): 329-342, 2023 04.
Article En | MEDLINE | ID: mdl-36828711

Tumors evolve together with the tumor microenvironment (TME) and reshape it towards immunosuppression. Immunostimulating cytokines can be used to revert this state leading to effective antitumor immune responses, but their exploitation as anticancer drugs has been hampered by severe toxicity associated with systemic administration. Local, TME-targeted delivery of immune activating cytokines can deploy their antitumoral function more effectively than systemic administration while, at the same time, avoiding exposure of healthy organs and limiting toxicity. Here, we review different gene and cell therapy platforms developed for tumor-directed cytokine delivery highlighting their potential for clinical translation.


Antineoplastic Agents , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Cytokines , Neoplasms/genetics , Neoplasms/therapy , Immunotherapy , Tumor Microenvironment/genetics
4.
Sci Transl Med ; 14(653): eabl4106, 2022 07 13.
Article En | MEDLINE | ID: mdl-35857642

Glioblastoma multiforme (GBM) is the most common and lethal brain tumor characterized by a strongly immunosuppressive tumor microenvironment (TME) that represents a barrier also for the development of effective immunotherapies. The possibility to revert this hostile TME by immunoactivating cytokines is hampered by the severe toxicity associated with their systemic administration. Here, we exploited a lentiviral vector-based platform to engineer hematopoietic stem cells ex vivo with the aim of releasing, via their tumor-infiltrating monocyte/macrophage progeny, interferon-α (IFN-α) or interleukin-12 (IL-12) at the tumor site with spatial and temporal selectivity. Taking advantage of a syngeneic GBM mouse model, we showed that inducible release of IFN-α within the TME achieved robust tumor inhibition up to eradication and outperformed systemic treatment with the recombinant protein in terms of efficacy, tolerability, and specificity. Single-cell RNA sequencing of the tumor immune infiltrate revealed reprogramming of the immune microenvironment toward a proinflammatory and antitumoral state associated with loss of a macrophage subpopulation shown to be associated with poor prognosis in human GBM. The spatial and temporal control of IL-12 release was critical to overcome an otherwise lethal hematopoietic toxicity while allowing to fully exploit its antitumor activity. Overall, our findings demonstrate a potential therapeutic approach for GBM and set the bases for a recently launched first-in-human clinical trial in patients with GBM.


Brain Neoplasms , Glioblastoma , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cytokines , Disease Models, Animal , Glioblastoma/drug therapy , Interferon-alpha , Interleukin-12/therapeutic use , Mice , Tumor Microenvironment
5.
Cancers (Basel) ; 13(2)2021 Jan 10.
Article En | MEDLINE | ID: mdl-33435218

Glioblastomas (GBM) are the most aggressive tumors affecting the central nervous system in adults, causing death within, on average, 15 months after diagnosis. Immunocompetent in-vivo models that closely mirror human GBM are urgently needed for deciphering glioma biology and for the development of effective treatment options. The murine GBM cell lines currently available for engraftment in immunocompetent mice are not only exiguous but also inadequate in representing prominent characteristics of human GBM such as infiltrative behavior, necrotic areas, and pronounced tumor heterogeneity. Therefore, we generated a set of glioblastoma cell lines by repeated in vivo passaging of cells isolated from a neural stem cell-specific Pten/p53 double-knockout genetic mouse brain tumor model. Transcriptome and genome analyses of the cell lines revealed molecular heterogeneity comparable to that observed in human glioblastoma. Upon orthotopic transplantation into syngeneic hosts, they formed high-grade gliomas that faithfully recapitulated the histopathological features, invasiveness and immune cell infiltration characteristic of human glioblastoma. These features make our cell lines unique and useful tools to study multiple aspects of glioblastoma pathomechanism and to test novel treatments in an intact immune microenvironment.

6.
Open Biol ; 10(5): 190262, 2020 05.
Article En | MEDLINE | ID: mdl-32397871

The transcription factor HIF-1α is overexpressed in chronic lymphocytic leukaemia (CLL), where it promotes leukaemia progression by favouring the interaction of leukaemic cells with protective tissue microenvironments. Here, we tested the hypothesis that a pharmacological compound previously shown to inhibit HIF-1α may act as a chemosensitizer by interrupting protective microenvironmental interactions and exposing CLL cells to fludarabine-induced cytotoxicity. We found that the camptothecin-11 analogue EZN-2208 sensitizes CLL cells to fludarabine-induced apoptosis in cytoprotective in vitro cultures; in vivo EZN-2208 improves fludarabine responses, especially in early phases of leukaemia expansion, and exerts significant anti-leukaemia activity, thus suggesting that this or similar compounds may be considered as effective CLL therapeutic approaches.


Camptothecin/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Polyethylene Glycols/administration & dosage , Vidarabine/analogs & derivatives , Adult , Aged , Animals , Camptothecin/administration & dosage , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Female , Gene Expression Regulation, Leukemic/drug effects , Humans , Male , Mice , Middle Aged , Neoplasm Transplantation , Polyethylene Glycols/pharmacology , Treatment Outcome , Tumor Microenvironment , Vidarabine/administration & dosage , Vidarabine/pharmacology
7.
JCI Insight ; 2(4): e87380, 2017 02 23.
Article En | MEDLINE | ID: mdl-28239645

Elucidating the molecular basis of tumor metastasis is pivotal for eradicating cancer-related mortality. Triple-negative breast cancer (TNBC) encompasses a class of aggressive tumors characterized by high rates of recurrence and metastasis, as well as poor overall survival. Here, we find that the promyelocytic leukemia protein PML exerts a prometastatic function in TNBC that can be targeted by arsenic trioxide. We found that, in TNBC patients, constitutive HIF1A activity induces high expression of PML, along with a number of HIF1A target genes that promote metastasis at multiple levels. Intriguingly, PML controls the expression of these genes by binding to their regulatory regions along with HIF1A. This mechanism is specific to TNBC cells and does not occur in other subtypes of breast cancer where PML and prometastatic HIF1A target genes are underexpressed. As a consequence, PML promotes cell migration, invasion, and metastasis in TNBC cell and mouse models. Notably, pharmacological inhibition of PML with arsenic trioxide, a PML-degrading agent used to treat promyelocytic leukemia patients, delays tumor growth, impairs TNBC metastasis, and cooperates with chemotherapy by preventing metastatic dissemination. In conclusion, we report identification of a prometastatic pathway in TNBC and suggest clinical development toward the use of arsenic trioxide for TNBC patients.


Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Promyelocytic Leukemia Protein/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Movement , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , NIH 3T3 Cells , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Promyelocytic Leukemia Protein/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
8.
Proc Natl Acad Sci U S A ; 113(41): E6219-E6227, 2016 10 11.
Article En | MEDLINE | ID: mdl-27671648

Cells in the tumor microenvironment may be reprogrammed by tumor-derived metabolites. Cholesterol-oxidized products, namely oxysterols, have been shown to favor tumor growth directly by promoting tumor cell growth and indirectly by dampening antitumor immune responses. However, the cellular and molecular mechanisms governing oxysterol generation within tumor microenvironments remain elusive. We recently showed that tumor-derived oxysterols recruit neutrophils endowed with protumoral activities, such as neoangiogenesis. Here, we show that hypoxia inducible factor-1a (HIF-1α) controls the overexpression of the enzyme Cyp46a1, which generates the oxysterol 24-hydroxycholesterol (24S-HC) in a pancreatic neuroendocrine tumor (pNET) model commonly used to study neoangiogenesis. The activation of the HIF-1α-24S-HC axis ultimately leads to the induction of the angiogenic switch through the positioning of proangiogenic neutrophils in proximity to Cyp46a1+ islets. Pharmacologic blockade or genetic inactivation of oxysterols controls pNET tumorigenesis by dampening the 24S-HC-neutrophil axis. Finally, we show that in some human pNET samples Cyp46a1 transcripts are overexpressed, which correlate with the HIF-1α target VEGF and with tumor diameter. This study reveals a layer in the angiogenic switch of pNETs and identifies a therapeutic target for pNET patients.


Cell Transformation, Neoplastic/metabolism , Hydroxycholesterols/metabolism , Neuroendocrine Tumors/etiology , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cholestanetriol 26-Monooxygenase/genetics , Cholestanetriol 26-Monooxygenase/metabolism , Cholesterol 24-Hydroxylase , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Enzyme Activation , Female , Fluorescent Antibody Technique , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Male , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Oncotarget ; 7(33): 53540-53557, 2016 Aug 16.
Article En | MEDLINE | ID: mdl-27447550

Hypoxia inducible transcription factors (HIFs) are the main regulators of adaptive responses to hypoxia and are often activated in solid tumors, but their role in leukemia is less clear. In acute myeloid leukemia (AML), in particular, controversial new findings indicate that HIF-1α can act either as an oncogene or a tumor suppressor gene, and this may depend on the stage of leukemia development and/or the AML sub-type.In this study, we find that HIF-1α promotes leukemia progression in the acute monocytic leukemia sub-type of AML through activation of an invasive phenotype. By applying a list of validated HIF-1α-target genes to different AML sub-types, we identified a HIF-1α signature that typifies acute monocytic leukemia when compared with all other AML sub-types. We validated expression of this signature in cell lines and primary cells from AML patients. Interestingly, this signature is enriched for genes that control cell motility at different levels. As a consequence, inhibiting HIF-1α impaired leukemia cell migration, chemotaxis, invasion and transendothelial migration in vitro, and this resulted in impaired bone marrow homing and leukemia progression in vivo. Our data suggest that in acute monocytic leukemia an active HIF-1α-dependent pro-invasive pathway mediates the ability of leukemic cells to migrate and invade extramedullary sites and may be targeted to reduce leukemia dissemination.


Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukemia, Monocytic, Acute/pathology , Animals , Cell Line, Tumor , Cell Movement/physiology , Disease Progression , Heterografts , Humans , Leukemia, Monocytic, Acute/metabolism , Mice , Phenotype
10.
Blood ; 127(16): 1987-97, 2016 Apr 21.
Article En | MEDLINE | ID: mdl-26825709

Hypoxia-inducible transcription factors (HIFs) regulate a wide array of adaptive responses to hypoxia and are often activated in solid tumors and hematologic malignancies due to intratumoral hypoxia and emerging new layers of regulation. We found that in chronic lymphocytic leukemia (CLL), HIF-1α is a novel regulator of the interaction of CLL cells with protective leukemia microenvironments and, in turn, is regulated by this interaction in a positive feedback loop that promotes leukemia survival and propagation. Through unbiased microarray analysis, we found that in CLL cells, HIF-1α regulates the expression of important chemokine receptors and cell adhesion molecules that control the interaction of leukemic cells with bone marrow and spleen microenvironments. Inactivation of HIF-1α impairs chemotaxis and cell adhesion to stroma, reduces bone marrow and spleen colonization in xenograft and allograft CLL mouse models, and prolongs survival in mice. Of interest, we found that in CLL cells, HIF-1α is transcriptionally regulated after coculture with stromal cells. Furthermore, HIF-1α messenger RNA levels vary significantly within CLL patients and correlate with the expression of HIF-1α target genes, including CXCR4, thus further emphasizing the relevance of HIF-1α expression to CLL pathogenesis.


Cell Communication/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Tumor Microenvironment/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Adhesion/genetics , Chemotaxis, Leukocyte/genetics , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/metabolism , Spleen/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
11.
Clin Cancer Res ; 21(16): 3685-94, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-25931453

PURPOSE: Retinoic acid-arsenic trioxide (ATRA-ATO) combination therapy is the current standard of care for patients with acute promyelocytic leukemia (APL) carrying the oncogenic fusion protein PML-RARα. Despite the high cure rates obtained with this drug combination, resistance to arsenic is recently emerging. Moreover, patients with APL carrying the PLZF-RARα fusion protein are partially resistant to ATRA treatment. Hypoxia-inducible factor-1α (HIF-1α) activation has been recently reported in APL, and EZN-2208 (PEG-SN38) is a compound with HIF-1α inhibitory function currently tested in clinical trials. This study investigates the effect of EZN-2208 in different preclinical APL models, either alone or in combination with ATRA. EXPERIMENTAL DESIGN: Efficacy of EZN-2208 in APL was measured in vitro by assessing expression of HIF-1α target genes, cell migration, clonogenicity, and differentiation, vis a vis the cytotoxic and cytostatic effects of this compound. In vivo, EZN-2208 was used in mouse models of APL driven by PML-RARα or PLZF-RARα, either alone or in combination with ATRA. RESULTS: Treatment of APL cell lines with noncytotoxic doses of EZN-2208 causes dose-dependent downregulation of HIF-1α bona fide target genes and affects cell migration and clonogenicity in methylcellulose. In vivo, EZN-2208 impairs leukemia progression and prolongs mice survival in APL mouse models. More importantly, when used in combination with ATRA, EZN-2208 synergizes in debulking leukemia and eradicating leukemia-initiating cells. CONCLUSIONS: Our preclinical data suggest that the combination ATRA-EZN-2208 may be tested to treat patients with APL who develop resistance to ATO or patients carrying the PLZF-RARα fusion protein.


Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Leukemia, Promyelocytic, Acute/drug therapy , Oncogene Proteins, Fusion/genetics , Tretinoin/administration & dosage , Animals , Arsenic Trioxide , Arsenicals/administration & dosage , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Differentiation/drug effects , Cell Movement/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Mice , Mutation , Neoplasm Proteins/biosynthesis , Oxides/administration & dosage , Polyethylene Glycols/administration & dosage
12.
Genome Med ; 6(12): 84, 2014.
Article En | MEDLINE | ID: mdl-25452766

BACKGROUND: Acute promyelocytic leukemia (APL) is a sub-type of acute myeloid leukemia (AML) characterized by a block of myeloid differentiation at the promyelocytic stage and the predominant t(15:17) chromosomal translocation. We have previously determined that cells from APL patients show increased expression of genes regulated by hypoxia-inducible transcription factors (HIFs) compared to normal promyelocytes. HIFs regulate crucial aspects of solid tumor progression and are currently being implicated in leukemogenesis. METHODS: To investigate the contribution of hypoxia-related signaling in APL compared to other AML sub-types, we reverse engineered a transcriptional network from gene expression profiles of AML patients' samples, starting from a list of direct target genes of HIF-1. A HIF-1-dependent subnetwork of genes specifically dysregulated in APL was derived from the comparison between APL and other AMLs. RESULTS: Interestingly, this subnetwork shows a unique involvement of genes related to extracellular matrix interaction and cell migration, with decreased expression of genes involved in cell adhesion and increased expression of genes implicated in motility and invasion, thus unveiling the presence of characteristics of epithelial-mesenchymal transition (EMT). We observed that the genes of this subnetwork, whose dysregulation shows a peculiar pattern across different AML sub-types, distinguish malignant from normal promyelocytes, thus ruling out dependence on a myeloid developmental stage. Also, expression of these genes is reversed upon treatment of APL-derived NB4 cells with all-trans retinoic acid and cell differentiation. CONCLUSIONS: Our data suggest that pathways related to EMT-like processes can be implicated also in hematological malignancies besides solid tumors, and can identify specific AML sub-types.

13.
Stem Cell Reports ; 2(6): 794-809, 2014 Jun 03.
Article En | MEDLINE | ID: mdl-24936467

Maintenance and differentiation of hematopoietic stem cells (HSCs) is regulated through cell-autonomous and non-cell-autonomous mechanisms within specialized bone marrow microenvironments. Recent evidence demonstrates that signaling by HIF-1α contributes to cell-autonomous regulation of HSC maintenance. By investigating the role of HIF factors in bone marrow mesenchymal progenitors, we found that murine endosteal mesenchymal progenitors express high levels of HIF-1α and HIF-2α and proliferate preferentially in hypoxic conditions ex vivo. Inactivation of either HIF-1α or HIF-2α dramatically affects their phenotype, propagation, and differentiation. Also, downregulation of HIF factors provokes an increase in interferon-responsive genes and triggers expansion and differentiation of hematopoietic progenitors by a STAT1-mediated mechanism. Interestingly, in conditions of demand-driven hematopoiesis HIF factors are specifically downregulated in mesenchymal progenitors in vivo. In conclusion, our findings indicate that HIF factors also regulate hematopoiesis non-cell-autonomously by preventing activation of a latent program in mesenchymal progenitors that promotes hematopoiesis.


Bone Marrow Cells/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Hematopoiesis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mesenchymal Stem Cells/cytology , Mice , Signal Transduction/physiology
14.
EMBO Mol Med ; 6(5): 640-50, 2014 May.
Article En | MEDLINE | ID: mdl-24711541

Acute promyelocytic leukemia (APL) is epitomized by the chromosomal translocation t(15;17) and the resulting oncogenic fusion protein PML-RARα. Although acting primarily as a transcriptional repressor, PML-RARα can also exert functions of transcriptional co-activation. Here, we find that PML-RARα stimulates transcription driven by HIF factors, which are critical regulators of adaptive responses to hypoxia and stem cell maintenance. Consistently, HIF-related gene signatures are upregulated in leukemic promyelocytes from APL patients compared to normal promyelocytes. Through in vitro and in vivo studies, we find that PML-RARα exploits a number of HIF-1α-regulated pro-leukemogenic functions that include cell migration, bone marrow (BM) neo-angiogenesis and self-renewal of APL blasts. Furthermore, HIF-1α levels increase upon treatment of APL cells with all-trans retinoic acid (ATRA). As a consequence, inhibiting HIF-1α in APL mouse models delays leukemia progression and exquisitely synergizes with ATRA to eliminate leukemia-initiating cells (LICs).


Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukemia, Promyelocytic, Acute/physiopathology , Nuclear Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Physiological Phenomena , Disease Models, Animal , Humans , Mice , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recurrence , Retinoic Acid Receptor alpha , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
15.
EMBO Mol Med ; 3(5): 249-57, 2011 May.
Article En | MEDLINE | ID: mdl-21387562

The promyelocytic leukaemia gene PML is a pleiotropic tumour suppressor. We have recently demonstrated that PML opposes mTOR-HIF1α-VEGF signalling in hypoxia. To determine the relevance of PML-mTOR antagonism in tumourigenesis, we have intercrossed Pml null mice with Tsc2 heterozygous mice, which develop kidney cysts and carcinomas exhibiting mTOR upregulation. We find that combined inactivation of Pml and Tsc2 results in aberrant TORC1 activity both in pre-tumoural kidneys as well as in kidney lesions. Such increase correlates with a marked acceleration in tumour progression, impacting on both the biology and histology of kidney carcinomas. Also, Pml inactivation decreases the rate of loss of heterozygosity (LOH) for the wt Tsc2 allele. Interestingly, however, aberrant TORC1 activity does not accelerate renal cystogenesis in Tsc2/Pml mutants. Our data demonstrate that activation of mTOR is critical for tumour progression, but not for tumour initiation in the kidney.


Gene Expression Regulation , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , TOR Serine-Threonine Kinases/biosynthesis , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma/pathology , Crosses, Genetic , Histocytochemistry , Immunohistochemistry , Kidney/pathology , Kidney Neoplasms/pathology , Magnetic Resonance Imaging , Mice , Mice, Knockout , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Radiography, Abdominal , Transcription Factors/biosynthesis , Transcription Factors/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
16.
FASEB J ; 24(8): 2680-8, 2010 Aug.
Article En | MEDLINE | ID: mdl-20354140

Loss of the fumarate hydratase (FH) tumor suppressor gene results in the development of benign tumors that rarely, but regrettably, progress to very aggressive cancers. Using mouse embryo fibroblasts (MEFs) to model transformation, we found that fh knockdown results in increased expression of the met oncogene-encoded tyrosine kinase receptor through hypoxia-inducible factor (hif) stabilization. MET-increased expression was alone able to stabilize hif, thus establishing a feed forward loop that might enforce tumor progression. The fh-defective MEFs showed increased motility and protection from apoptosis. Motility, but not survival, relied on hif-1alpha and was greatly enhanced by MET ligand hepatocyte growth factor. Met cooperated with a weakly oncogenic ras in making MEFs transformed and tumorigenic, as shown by in vitro and in vivo assays. Loss of fh was not equally effective by itself but enhanced the transformed and tumorigenic phenotype induced by ras and MET. Consistently, the rescue of fumarase expression abrogated the motogenic and transformed phenotype of fh-defective MEFs. In conclusion, the data suggest that the progression of tumors where FH is lost might be boosted by activation of the MET oncogene, which is able to drive cell-autonomous tumor progression and is a strong candidate for targeted therapy.


Cell Transformation, Neoplastic , Fumarate Hydratase/physiology , Neoplasms/etiology , Proto-Oncogene Proteins c-met/physiology , Animals , Cells, Cultured , Fibroblasts , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Hepatocyte Growth Factor/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Tumor Suppressor Proteins/genetics
17.
Clin Cancer Res ; 13(7): 2191-8, 2007 Apr 01.
Article En | MEDLINE | ID: mdl-17404103

PURPOSE: Advanced ovarian cancers are initially responsive to combinatorial chemotherapy with platinum drugs and taxanes but, in most cases, develop drug resistance. We recently showed that, in vitro, hepatocyte growth factor (HGF) enhances death of human ovarian cancer cell lines treated with cisplatin (CDDP) and paclitaxel. The present study addresses whether in vivo HGF makes ovarian carcinoma cells more responsive to these chemotherapeutics. EXPERIMENTAL DESIGN: Using Lentiviral vectors carrying the HGF transgene, we transduced SK-OV-3 and NIH:OVCAR-3 ovarian carcinoma cell lines to obtain stable autocrine and paracrine HGF receptor activation. In vitro, we assayed growth, motility, invasiveness, and the response to CDDP and paclitaxel of the HGF-secreting bulk unselected cell populations. In vivo, we tested the cytotoxic effects of the drugs versus s.c. tumors formed by the wild-type and HGF-secreting cells in immunocompromised mice. Tumor-bearing mice were treated with CDDP (i.p.) and paclitaxel (i.v.), combined in different schedules and doses. RESULTS: In vitro, HGF-secreting cells did not show altered proliferation rates and survival but were strongly sensitized to the death triggered by CDDP and paclitaxel, alone or in combination. In vivo, we found a therapeutic window in which autocrine/paracrine HGF made tumors sensitive to low doses of the drugs, which were ineffective on their own. CONCLUSIONS: These data provide the proof-of-concept that in vivo gene therapy with HGF might be competent in sensitizing ovarian cancer cells to conventional chemotherapy.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Genetic Therapy/methods , Hepatocyte Growth Factor/genetics , Ovarian Neoplasms/drug therapy , Animals , Apoptosis , Blotting, Western , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Flow Cytometry , Genetic Vectors , Hepatocyte Growth Factor/metabolism , Humans , In Vitro Techniques , Lentivirus/genetics , Mice , Ovarian Neoplasms/metabolism , Paclitaxel/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Transgenes
18.
Biochem Biophys Res Commun ; 347(3): 781-90, 2006 Sep 01.
Article En | MEDLINE | ID: mdl-16843435

Doxorubicin is the anthracycline with the widest spectrum of antitumor activity, and it has been shown that the antitumor activity is mediated in vivo by selective triggering of apoptosis in proliferating endothelial cells. We studied cultured human endothelial cells and observed that doxorubicin-induced apoptosis was mediated by p38 mitogen-activated protein kinase (MAPK). Doxorubicin-provoked apoptosis was significantly inhibited by expression of dominant negative p38 MAPK or pharmacological inhibition with SB203580. Furthermore, blocking phosphatidylinositol-3-kinase/Akt signaling significantly increased doxorubicin-induced caspase-3 activity and cell death, indicating that Akt is a survival factor in this system. Notably, we also found that doxorubicin-provoked apoptosis included p38 MAPK-mediated inhibition of Akt and Bad phosphorylation. Furthermore, doxorubicin-stimulated phosphorylation of Bad in cells expressing dominant negative p38 MAPK was impeded by the inhibition of PI3-K. In addition to the impact on Bad phosphorylation, doxorubicin-treatment caused p38 MAPK-dependent downregulation of Bcl-xL protein.


Apoptosis/drug effects , Down-Regulation , Doxorubicin/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , bcl-Associated Death Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Caspase 3 , Caspases/metabolism , Cell Line , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Humans , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , bcl-X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
19.
Mol Cancer Ther ; 5(5): 1126-35, 2006 May.
Article En | MEDLINE | ID: mdl-16731744

Advanced ovarian cancers are initially responsive to chemotherapy with platinum drugs but develop drug resistance in most cases. We showed recently that hepatocyte growth factor (HGF) enhances death of human ovarian cancer cell lines treated with cisplatin (CDDP) and that this effect is mediated by the p38 mitogen-activated protein kinase. In this work, we integrated genome-wide expression profiling, in silico data survey, and functional assays to identify transcripts regulated in SK-OV-3 ovarian cancer cells made more responsive to CDDP by HGF. Using oligonucleotide microarrays, we found that HGF pretreatment changes the transcriptional response to CDDP. Quantitative reverse transcription-PCR not only validated all the 15 most differentially expressed genes but also confirmed that they were primarily modulated by the combined treatment with HGF and CDDP and reversed by suppressing p38 mitogen-activated protein kinase activity. Among the differentially expressed genes, we focused functional analysis on two regulatory subunits of the protein phosphatase 2A, which were down-modulated by HGF plus CDDP. Decrease of each subunit by RNA interference made ovarian cancer cells more responsive to CDDP, mimicking the effect of HGF. In conclusion, we show that HGF and CDDP modulate transcription in ovarian cancer cells and that this transcriptional response is involved in apoptosis regulation. We also provide the proof-of-concept that the identified genes might be targeted to either increase the efficacy of chemotherapeutics or revert chemotherapy resistance.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/pharmacology , Ovarian Neoplasms/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Growth Factor/genetics , Humans , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2 , Protein Subunits/metabolism , Time Factors , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Cell Signal ; 18(11): 1967-76, 2006 Nov.
Article En | MEDLINE | ID: mdl-16677802

Hepatocyte growth factor (HGF) induces invasive growth, a biological program that confers tumor cells the capability to invade and metastasize by integrating cell proliferation, motility, morphogenesis, and survival. We here demonstrate that HGFR activation promotes survival of colorectal carcinoma (CRC) cells exposed to conditions that mimic those met during tumor progression, i.e. nutrient deprivation or substrate detachment, and following chemotherapeutic treatment. In all these conditions, a sustained activation of p38 MAPK delivers a main death signal that is overcome by cell treatment with HGF. HGF-driven survival requires the engagement of the PI3K/Akt/mTOR/p70S6K and ERK MAPK transduction pathways. Abrogation of p38 MAPK activity prevents CRC cell apoptosis also when these transduction pathways are inhibited, and treatment with HGF further increases survival. Engagement of these signaling cascades is also needed for HGF to induce CRC cell scattering, morphogenesis, motility and invasion. Activation of p38 MAPK signaling is therefore a main apoptotic switch for CRC cells in the stressful conditions encountered during tumor progression. Conversely, HGF orchestrates several biochemical pathways, which allow cell survival in these same conditions and promote the biological responses required for tumor invasive growth. Both p38 MAPK and HGF/HGFR signaling constitute potential molecular targets for inhibiting colorectal carcinogenesis.


Apoptosis , Colorectal Neoplasms/physiopathology , Hepatocyte Growth Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Hepatocyte Growth Factor/pharmacology , Humans , Neoplasm Invasiveness/physiopathology , Signal Transduction/drug effects
...