Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 108
1.
J Intern Med ; 276(1): 61-70, 2014 Jul.
Article En | MEDLINE | ID: mdl-24635409

The recent finding that almost all patients with acute promyelocytic leukaemia (APL) may be cured using a combination of retinoic acid (RA) and arsenic trioxide (As(2)O(3)) (N Engl J Med, 369, 2013 and 111) highlights the progress made in our understanding of APL pathogenesis and therapeutic approaches over the past 25 years. The study of APL has revealed many important lessons related to transcriptional control, nuclear organization, epigenetics and the role of proteolysis in biological control. Even more important has been the clinical demonstration that molecularly targeted therapy can eradicate disease.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Molecular Targeted Therapy/methods , Arsenic Trioxide , Arsenicals/administration & dosage , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic/physiology , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Oxides/administration & dosage , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptor alpha/metabolism , Signal Transduction/physiology , Tretinoin/administration & dosage
2.
Br J Cancer ; 110(6): 1413-9, 2014 Mar 18.
Article En | MEDLINE | ID: mdl-24569467

BACKGROUND: Triple-negative (TN) breast cancers exhibit major initial responses to neoadjuvant chemotherapy, but generally have a poor outcome. Because of the lack of validated drug targets, chemotherapy remains an important therapeutic tool in these cancers. METHODS: We report the survival of two consecutive series of 267 locally advanced breast cancers (LABC) treated with two different neoadjuvant regimens, either a dose-dense and dose-intense cyclophosphamide-anthracycline (AC) association (historically called SIM) or a conventional sequential association of cyclophosphamide and anthracycline, followed by taxanes (EC-T). We compared pathological responses and survival rates of these two groups and studied their association with tumours features. RESULTS: Although the two regimens showed equivalent pathological complete response (pCR) in the whole population (16 and 12%), the SIM regimen yielded a non-statistically higher pCR rate than EC-T (48% vs 24%, P=0.087) in TN tumours. In the SIM protocol, DFS was statistically higher for TN than for non-TN patients (P=0.019), although we showed that the TN status was associated with an increased initial risk of recurrence in both regimens. This effect gradually decreased and after 2 years, TN was associated with a significantly decreased likelihood of relapse in SIM-treated LABC (hazard ratio (HR)=0.25 (95% CI: 0.07-0.86), P=0.028). CONCLUSIONS: AC dose intensification treatment is associated with a very favourable long-term survival rate in TN breast cancers. These observations call for a prospective assessment of such dose-intense AC-based regimens in locally advanced TN tumours.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Adult , Aged , Cyclophosphamide/administration & dosage , Disease-Free Survival , Dose-Response Relationship, Drug , Epirubicin/administration & dosage , Female , Humans , Middle Aged , Neoadjuvant Therapy , Prognosis , Prospective Studies , Survival Rate , Survivors , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/surgery , Young Adult
3.
Leukemia ; 26(7): 1630-7, 2012 Jul.
Article En | MEDLINE | ID: mdl-22333881

Aberrant histone acetylation was physiopathologically associated with the development of acute myeloid leukemias (AMLs). Reversal of histone deacetylation by histone deacetylase inhibitor (HDACis) activates a cell death program that allows tumor regression in mouse models of AMLs. We have used several models of PML-RARA-driven acute promyelocytic leukemias (APLs) to analyze the in vivo effects of valproic acid, a well-characterized HDACis. Valproic acid (VPA)-induced rapid tumor regression and sharply prolonged survival. However, discontinuation of treatment was associated to an immediate relapse. In vivo, as well as ex vivo, VPA-induced terminal granulocytic differentiation. Yet, despite full differentiation, leukemia-initiating cell (LIC) activity was actually enhanced by VPA treatment. In contrast to all-trans retinoic acid (ATRA) or arsenic, VPA did not degrade PML-RARA. However, in combination with ATRA, VPA synergized for PML-RARA degradation and LIC eradication in vivo. Our studies indicate that VPA triggers differentiation, but spares LIC activity, further uncouple differentiation from APL clearance and stress the importance of PML-RARA degradation in APL cure.


Anticonvulsants/pharmacology , Cell Differentiation/drug effects , Cell Transformation, Neoplastic/drug effects , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/pathology , Valproic Acid/pharmacology , Acetylation/drug effects , Animals , Antineoplastic Agents/pharmacology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Recurrence, Local , Oncogene Proteins, Fusion/metabolism , Signal Transduction , Tretinoin/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Leukemia ; 26(3): 451-60, 2012 Mar.
Article En | MEDLINE | ID: mdl-21869839

The vitamin E derivative (+)α-tocopheryl succinate (α-TOS) exerts pro-apoptotic effects in a wide range of tumors and is well tolerated by normal tissues. Previous studies point to a mitochondrial involvement in the action mechanism; however, the early steps have not been fully elucidated. In a model of acute promyelocytic leukemia (APL) derived from hCG-PML-RARα transgenic mice, we demonstrated that α-TOS is as effective as arsenic trioxide or all-trans retinoic acid, the current gold standards of therapy. We also demonstrated that α-TOS induces an early dissipation of the mitochondrial membrane potential in APL cells and studies with isolated mitochondria revealed that this action may result from the inhibition of mitochondrial respiratory chain complex I. Moreover, α-TOS promoted accumulation of reactive oxygen species hours before mitochondrial cytochrome c release and caspases activation. Therefore, an in vivo antileukemic action and a novel mitochondrial target were revealed for α-TOS, as well as mitochondrial respiratory complex I was highlighted as potential target for anticancer therapy.


Arsenicals/therapeutic use , Electron Transport Complex I/antagonists & inhibitors , Leukemia, Promyelocytic, Acute/drug therapy , Mitochondria/drug effects , Oxides/therapeutic use , Tretinoin/therapeutic use , alpha-Tocopherol/pharmacology , alpha-Tocopherol/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Arsenic Trioxide , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Disease Models, Animal , Electron Transport Complex II/antagonists & inhibitors , Humans , Leukemia, Promyelocytic, Acute/mortality , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Stability/drug effects , Rats , Reactive Oxygen Species/metabolism , Transplantation, Isogeneic
5.
Oncogene ; 31(9): 1196-206, 2012 Mar 01.
Article En | MEDLINE | ID: mdl-21785460

The current histoclinical breast cancer classification is simple but imprecise. Several molecular classifications of breast cancers based on expression profiling have been proposed as alternatives. However, their reliability and clinical utility have been repeatedly questioned, notably because most of them were derived from relatively small initial patient populations. We analyzed the transcriptomes of 537 breast tumors using three unsupervised classification methods. A core subset of 355 tumors was assigned to six clusters by all three methods. These six subgroups overlapped with previously defined molecular classes of breast cancer, but also showed important differences, notably the absence of an ERBB2 subgroup and the division of the large luminal ER+ group into four subgroups, two of them being highly proliferative. Of the six subgroups, four were ER+/PR+/AR+, one was ER-/PR-/AR+ and one was triple negative (AR-/ER-/PR-). ERBB2-amplified tumors were split between the ER-/PR-/AR+ subgroup and the highly proliferative ER+ LumC subgroup. Importantly, each of these six molecular subgroups showed specific copy-number alterations. Gene expression changes were correlated to specific signaling pathways. Each of these six subgroups showed very significant differences in tumor grade, metastatic sites, relapse-free survival or response to chemotherapy. All these findings were validated on large external datasets including more than 3000 tumors. Our data thus indicate that these six molecular subgroups represent well-defined clinico-biological entities of breast cancer. Their identification should facilitate the detection of novel prognostic factors or therapeutical targets in breast cancer.


Breast Neoplasms/classification , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cluster Analysis , Databases, Genetic , Female , Gene Expression Profiling , Humans , Prognosis , Reproducibility of Results , Signal Transduction , Survival Analysis , Transcriptome , Treatment Outcome
6.
Br J Cancer ; 104(11): 1739-46, 2011 May 24.
Article En | MEDLINE | ID: mdl-21540864

BACKGROUND: Immunohistochemistry (IHC) and fluorescent in situ hybridisation (FISH) are currently the most commonly used methods to assess HER2 status. PCR-based assays allow quantitative determination of HER2 amplification (Q-PCR) or overexpression (Q-RT-PCR), but are not routinely used. We evaluated the relevance of Q-RT-PCR for HER2 status determination. METHODS: We compared IHC and Q-RT-PCR in 466 breast tumours. In discordant or equivocal cases, five additional methods (IHC with two other antibodies, FISH, silver in situ hybridisation (SISH) and Q-PCR) were combined to determine HER2 status. Two cases with HER2 intra-tumour heterogeneity were further explored by allelic profiles analysis and HUMARA clonality determination after microdissection. RESULTS: We observed 97.3% concordance between Q-RT-PCR and non-equivocal IHC. Twelve out of 466 cases (3%) revealed discordances between the two methods. The power of Q-RT-PCR to predict HER2 status (defined by seven methods) was similar to that of IHC. Although rare, some discordances between techniques might be due to HER2 intra-tumour heterogeneity and we report two examples, one tumour containing two distinct clones, another tumour consisting of HER2 amplified and non-amplified subclones. CONCLUSION: Q-RT-PCR and IHC are highly concordant methods for HER2 status assessment, and Q-RT-PCR allows a highly reliable quantitative assessment and could be a useful adjunct to IHC.


Reverse Transcriptase Polymerase Chain Reaction/methods , Alleles , Gene Dosage , Genes, erbB-2 , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Receptors, Androgen
7.
Cell Death Differ ; 16(2): 298-311, 2009 Feb.
Article En | MEDLINE | ID: mdl-19023333

Promyelomonocytic leukemia (PML) is a prominent oncosuppressor whose inactivation is involved in the pathogenesis of hematological and epithelial cancers. Here, we report that PML aggregated in nuclear bodies in syncytia elicited by the envelope glycoprotein complex (Env) of human immunodeficiency virus-1 (HIV-1) in vitro. PML aggregation occurred after the fusion of nuclei (karyogamy) within syncytia but before the apoptotic program was activated. The aggregation of PML was detectable in syncytia present in the brain or lymph nodes from patients with HIV-1 infection, as well as in a fraction of blood leukocytes, correlating with viral status. Using a range of specific inhibitors of PML (the oncogenic PML/RARalpha fusion product or specific small interfering RNAs), we demonstrated that, in Env-elicited syncytia, PML was required for activating phosphorylation of ataxia telangiectasia mutated (ATM), which colocalized with PML in nuclear bodies, in a molecular complex that also involved topoisomerase IIbeta-binding protein 1. PML knockdown thus inhibited the ATM-dependent DNA damage response that culminates in the activation of p53, p53-dependent transcription of pro-apoptotic genes and cell death. Infection of CD4-expressing cells with HIV-1 also induced syncytial apoptosis, which could be suppressed by inhibiting PML. Altogether, these data indicate that PML activation is a critical early event that participates in the apoptotic demise of HIV-1-elicited syncytia.


Apoptosis , HIV-1 , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , env Gene Products, Human Immunodeficiency Virus/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Giant Cells/virology , HeLa Cells , Humans , Promyelocytic Leukemia Protein , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Signal Transduction , Tumor Suppressor Protein p53/metabolism
8.
Leukemia ; 22(6): 1125-30, 2008 Jun.
Article En | MEDLINE | ID: mdl-18354491

In acute promyelocytic leukemia (APL), all-trans retinoic acid (ATRA) triggers cell differentiation, while arsenic trioxide (As(2)O(3)) generates partial differentiation and apoptosis. Animal and human studies suggest that newly diagnosed APL patients can be cured using As(2)O(3) combined with ATRA. Cyclooxygenases are involved in prostaglandins and thromboxane synthesis. We have recently demonstrated that ATRA induces cyclooxygenase-1 (COX-1) expression and prostaglandin synthesis in NB4 cells and in blasts from patients with APL. In the present study we investigated the effect of ATRA and As(2)O(3) co-treatment on COX-1 expression and prostaglandin formation and tested the effect of the COX-1/COX-2 nonselective inhibitor indomethacin on cell differentiation. Arsenic treatment of NB4 cells resulted in a partial but significant reduction of ATRA-dependent induction of COX-1 expression and activity. Pretreatment of NB4 cells with indomethacin significantly impaired ATRA/As(2)O(3)-induced differentiation, as assessed by cell morphology, nitroblue tetrazolium test or CD11c expression. PGE(2) reversed the negative effect of indomethacin on differentiation of ATRA/As(2)O(3)-treated NB4 cells. In conclusion, COX-1 contributes to ATRA-dependent maturation of NB4 cells and is affected by As(2)O(3). These results also suggest that nonsteroidal antiinflammatory drugs should be avoided in APL patients treated with the combination of ATRA and As(2)O(3).


Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Cyclooxygenase 1/metabolism , Dinoprostone/metabolism , Leukemia, Promyelocytic, Acute/drug therapy , Oxides/pharmacology , Tretinoin/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Arsenic Trioxide , Blotting, Western , CD11c Antigen/metabolism , Cell Differentiation , Drug Therapy, Combination , Humans , Indomethacin/pharmacology , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Models, Biological , Tretinoin/pharmacology , Tumor Cells, Cultured
9.
Oncogene ; 27(12): 1665-76, 2008 Mar 13.
Article En | MEDLINE | ID: mdl-17891179

Constitutive activation of the NF-kappaB pathway by the Tax oncoprotein plays a crucial role in the proliferation and transformation of HTLV-I infected T lymphocytes. We have previously shown that Tax ubiquitylation on C-terminal lysines is critical for binding of Tax to IkappaB kinase (IKK) and its subsequent activation. Here, we report that ubiquitylated Tax is not associated with active cytosolic IKK subunits, but binds endogenous IKK-alpha, -beta, -gamma, targeting them to the centrosome. K63-ubiquitylated Tax colocalizes at the centrosome with IKK-gamma, while K48-ubiquitylated Tax is stabilized upon proteasome inhibition. Altogether, these results support a model in which K63-ubiquitylated Tax activates IKK in a centrosome-associated signalosome, leading to the production of Tax-free active cytoplasmic IKK. These observations highlight an unsuspected link between Tax-induced IKK activation and the centrosome.


Centrosome/metabolism , Gene Products, tax/metabolism , I-kappa B Kinase/metabolism , Signal Transduction/physiology , Ubiquitin/metabolism , Carrier Proteins/metabolism , Cell Line, Transformed , Enzyme Activation/physiology , HeLa Cells , Human T-lymphotropic virus 1/metabolism , Humans , Protein Binding , Protein Subunits/metabolism , Transcriptional Elongation Factors
10.
Br J Cancer ; 97(8): 1157-64, 2007 Oct 22.
Article En | MEDLINE | ID: mdl-17876337

In advanced breast cancers, TP53 mutation is highly predictive of complete response to high-dose epirubicin/cyclophosphamide chemotherapy. In these tumours with an altered control of genomic stability, accumulation of chemotherapy-induced genetic alterations may contribute to cell death and account for complete response. To explore the effects of chemotherapy on stability of the tumour genome, allelic profiles were obtained from microdissected tumour samples before and after chemotherapy in 29 unresponsive breast cancers (9 with TP53 mutation). Ninety-four per cent allelic profiles remained unchanged after treatment. Interestingly, 11 profiles (6%) showed important changes after treatment; allelic imbalances significantly increased (four cases) or decreased (seven cases) after chemotherapy in three distinct experiments, two of which using laser microdissected tumour cells. These genetic changes were not linked to the TP53 status, but one tumour showed complete disappearance of TP53-mutated cells in the residual tumour after treatment. Altogether, these observations carry important implications for the clonal evolution of breast cancers treated with DNA-damaging agents, as they point both to the importance of tumour heterogeneity and chemotherapy-driven selection of subclones.


Allelic Imbalance/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Genes, p53/genetics , Adult , Aged , Aged, 80 and over , Cyclophosphamide/administration & dosage , Epirubicin/administration & dosage , Female , Humans , Lasers , Microdissection , Middle Aged , Mutation , Polymerase Chain Reaction
11.
Leukemia ; 21(8): 1792-801, 2007 Aug.
Article En | MEDLINE | ID: mdl-17568816

Kaposi's sarcoma (KS)-associated herpes virus (KSHV) is the causative agent of primary effusion lymphoma and of KS. Primary effusion lymphoma (PEL) is an aggressive proliferation of B cells. Conventional chemotherapy has limited benefits in PEL patients, and the prognosis is very poor. We previously reported that treatment of human T-cell leukemia virus type 1 (HTLV-1)-associated adult T-cell leukemia/lymphoma cells either with arsenic trioxide (As) combined to interferon-alpha (IFN-alpha) or with the bortezomib (PS-341) proteasome inhibitor induces cell cycle arrest and apoptosis, partly due to the reversal of the constitutive nuclear factor-kappaB (NF-kappaB) activation. PEL cells also display an activated NF-kappaB pathway that is necessary for their survival. This prompted us to investigate the effects of PS-341, or of the As/IFN-alpha combination on PEL cells. A dramatic inhibition of cell proliferation and induction of apoptosis was observed in PS-341 and in As/IFN-alpha treated cells. This was associated with the dissipation of the mitochondrial membrane potential, cytosolic release of cytochrome c, caspase activation and was reversed by the z-VAD caspase inhibitor. PS-341 and As/IFN-alpha treatment abrogated NF-kappaB translocation to the nucleus and decreased the levels of the anti-apoptotic protein Bcl-X(L). Altogether, these results provide a rational basis for a future therapeutic use of PS-341 or combined As and IFN-alpha in PEL patients.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Caspases/metabolism , Herpesvirus 8, Human/physiology , Lymphoma/pathology , Lymphoma/virology , Pyrazines/pharmacology , Arsenic Trioxide , Arsenicals/administration & dosage , Bortezomib , Cell Proliferation/drug effects , Humans , Interferon-alpha/administration & dosage , Lymphoma/enzymology , Membrane Potential, Mitochondrial/drug effects , NF-kappa B/metabolism , Oxides/administration & dosage , Protease Inhibitors/pharmacology , bcl-X Protein/metabolism
12.
Curr Top Microbiol Immunol ; 313: 129-44, 2007.
Article En | MEDLINE | ID: mdl-17217042

Arsenic has recently been identified as an effective drug in the treatment of newly diagnosed and relapsed acute promyelocytic leukemia. Indeed, arsenic trioxide combined with all-trans retinoic acid shows a synergistic effect. Mechanistically, arsenic targets the key leukemogenic protein PML-RARalpha, setting up a new example of molecular target-based cancer therapy.


Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Oxides/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Arsenic Trioxide , Arsenicals/administration & dosage , Arsenicals/adverse effects , Arsenicals/pharmacokinetics , Clinical Trials as Topic , Humans , Oxides/administration & dosage , Oxides/adverse effects , Oxides/pharmacokinetics , Treatment Outcome
14.
Leukemia ; 18(3): 607-15, 2004 Mar.
Article En | MEDLINE | ID: mdl-14712289

N-(4-hydroxyphenyl)retinamide (HPR) is a synthetic retinoid that inhibits growth and induces apoptosis in many human cell lines. We explored the effects of HPR on human T-cell lymphotropic virus type I (HTLV-I)-positive and HTLV-I-negative malignant T-cell lines, most of which are resistant to all-trans retinoic acid. Clinically achievable concentrations of HPR caused a dramatic inhibition of cell proliferation, G(0)/G(1) arrest, and massive apoptosis in all tested malignant T cells, while no effect was observed on resting or activated normal lymphocytes. Interestingly, HTLV-I-negative cell lines were significantly more sensitive to HPR compared to HTLV-I-positive and Tax-transfected cells. In HTLV-I-negative cells only, HPR-induced apoptosis was associated with ceramide accumulation, sharp decrease in mitochondrial membrane potential, and activation of caspases 8, 9 and 3, and could be partially reverted by the caspase inhibitor z-VAD suggesting that Tax protects infected cells from ceramide accumulation and caspase-mediated apoptosis. In HTLV-I-positive cells, HPR treatment rapidly induced proteasomal-mediated degradation of p21, downregulated cyclin D(1), and upregulated bax protein levels. These findings support a potential therapeutic role for HPR in both HTLV-I-associated adult T-cell leukemia/lymphoma (ATL) and HTLV-I-negative peripheral T-cell lymphomas.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Transformation, Viral , Fenretinide/pharmacology , Human T-lymphotropic virus 1/physiology , Proto-Oncogene Proteins c-bcl-2 , T-Lymphocytes/pathology , Caspases/metabolism , Cell Division/drug effects , Cell Line, Transformed , Cyclin D , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Humans , Proto-Oncogene Proteins/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Tumor Cells, Cultured , bcl-2-Associated X Protein
15.
Lancet ; 360(9336): 852-4, 2002 Sep 14.
Article En | MEDLINE | ID: mdl-12243922

TP53 activation by genotoxic drugs can induce apoptosis or cell-cycle arrest. Thus, whether the gene is mutated or wild type could affect the response of a tumour to chemotherapy. Clinical data are unclear, possibly as a result of heterogeneity of tumours, drugs, methods of assessing response, or TP53 status. We studied 50 non-inflammatory, locally advanced breast cancers that had been treated with high doses of a combination of epirubicin and cyclophosphamide. We noted eight complete responses, which all occurred in the 14 patients with tumours containing mutated TP53 (p<0.0001). In high-grade, advanced breast cancers, inactivation of the TP53 pathway could greatly improve the response to this chemotherapy regimen.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Death/genetics , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , DNA Mutational Analysis , Dose-Response Relationship, Drug , Drug Administration Schedule , Epirubicin/administration & dosage , Epirubicin/adverse effects , Female , Humans , Middle Aged , Neoplasm Staging , Prognosis , Treatment Outcome
17.
Oncogene ; 20(49): 7257-65, 2001 Oct 29.
Article En | MEDLINE | ID: mdl-11704854

Although there is evidence to suggest that PML/RARalpha expression is not the sole genetic event required for the development of acute promyelocytic leukemia (APL), there is little doubt that the fusion protein plays a central role in the initiation of leukemogenesis. The two therapeutic agents, retinoic acid and arsenic, that induce clinical remissions in APL, both target the oncogenic fusion protein, representing the first example of oncogene-directed cancer therapy. This review focuses on the molecular mechanisms accounting for PML/RARalpha degradation. Each drug targets a specific moiety of the fusion protein (RARalpha for retinoic acid, PML for arsenic) to the proteasome. Moreover, both activate a common caspase-dependent cleavage in the PML part of the fusion protein. Specific molecular determinants (the AF2 transactivator domain of RARalpha for retinoic acid and the K160 SUMO-binding site in PML for arsenic) are respectively implicated in RA- or arsenic-triggered catabolism. The respective roles of PML/RARalpha activation versus its catabolism are discussed with respect to differentiation or apoptosis induction in the context of single or dual therapies.


Arsenicals/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Oxides/pharmacology , Tretinoin/pharmacology , Animals , Arsenic Trioxide , Binding Sites , Caspases/metabolism , Cell Differentiation/drug effects , Cysteine Endopeptidases/metabolism , Humans , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex , Protein Structure, Tertiary , Remission Induction
18.
Lab Invest ; 81(10): 1397-402, 2001 Oct.
Article En | MEDLINE | ID: mdl-11598152

Solid tumors are composed not only of tumor cells but also of stromal nonneoplastic cells. In whole tumor samples, stromal cells retaining their alleles may therefore obscure detection of loss of heterozygosity (LOH) in tumor cells. An increasing number of studies have used laser-assisted tissue microdissection to improve LOH detection, but the real gain in sensitivity has been poorly quantified. We studied a group of 16 inflammatory breast carcinomas that were submitted to both standard DNA extraction from frozen whole tumor samples and laser microdissection performed on paraffin-embedded tumor samples. Using PCR with fluorescence-labeled primers, we comparatively analyzed ten polymorphic markers with both sources of DNA. With the LOH detection threshold set at -25%, we showed that 25 LOHs could not be diagnosed with whole tumor samples out of 73 LOHs positively diagnosed in microdissected samples (34%). With the LOH detection threshold set at -50%, the respective figures were 39 LOHs not diagnosed out of 55 LOHs (71%). Measuring the intensity of the allelic decrease, we showed that the mean decrease of the lost allele is -34% with whole tumor samples and -67% with microdissected samples. The increase in sensitivity of LOH detection with microdissection is associated with the density of stromal cells. This strong improvement in LOH detection in this aggressive type of breast cancer indicates that many other molecular studies performed on heterogeneous solid tumors may benefit from a first step of laser microdissection.


Breast Neoplasms/genetics , Loss of Heterozygosity , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Female , Humans , Inflammation , Lasers , Polymerase Chain Reaction , Tissue Embedding
19.
EMBO J ; 20(13): 3495-505, 2001 Jul 02.
Article En | MEDLINE | ID: mdl-11432836

The promyelocytic leukaemia (PML) protein localizes in the nucleus both in the nucleoplasm and in matrix-associated multiprotein complexes known as nuclear bodies (NBs). The number and the intensity of PML NBs increase in response to interferon (IFN). Overexpression of PML affects the replication of vesicular stomatitis virus and influenza virus. However, PML has a less powerful antiviral activity against these viruses than the IFN mediator MxA. Here, we show that overexpression of PML, but not that of Mx1 or MxA, leads to a drastic decrease of a complex retrovirus, the human foamy virus (HFV), gene expression. PML represses HFV transcription by complexing the HFV transactivator, Tas, preventing its direct binding to viral DNA. This physical interaction requires the N-terminal region of Tas and the RING finger of PML, but does not necessitate PML localization in NBs. Finally, we show that IFN treatment inhibits HFV replication in wild-type but not in PML-/- cells. These findings point to a role for PML in transcriptional repression and suggest that PML could play a key role in mediating an IFN-induced antiviral state against a complex retrovirus.


Antiviral Agents/metabolism , GTP-Binding Proteins , Interferon-alpha/pharmacology , Neoplasm Proteins/metabolism , Proteins/metabolism , Spumavirus/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Astrocytoma , CHO Cells , Cricetinae , DNA, Viral/genetics , Fibroblasts/physiology , Fibroblasts/virology , Glioblastoma , Humans , L Cells , Mice , Mice, Knockout , Myxovirus Resistance Proteins , Neoplasm Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , RNA, Messenger/genetics , RNA, Viral/genetics , RNA-Directed DNA Polymerase/metabolism , Recombinant Proteins/metabolism , Spumavirus/drug effects , Spumavirus/genetics , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured , Tumor Suppressor Proteins , Virus Replication/drug effects
20.
J Exp Med ; 193(12): 1361-71, 2001 Jun 18.
Article En | MEDLINE | ID: mdl-11413191

Promyelocytic leukemia (PML) is the organizer of nuclear matrix domains, PML nuclear bodies (NBs), with a proposed role in apoptosis control. In acute promyelocytic leukemia, PML/retinoic acid receptor (RAR) alpha expression disrupts NBs, but therapies such as retinoic acid or arsenic trioxide (As2O3) restore them. PML is conjugated by the ubiquitin-related peptide SUMO-1, a process enhanced by As2O3 and proposed to target PML to the nuclear matrix. We demonstrate that As2O3 triggers the proteasome-dependent degradation of PML and PML/RARalpha and that this process requires a specific sumolation site in PML, K160. PML sumolation is dispensable for its As2O3-induced matrix targeting and formation of primary nuclear aggregates, but is required for the formation of secondary shell-like NBs. Interestingly, only these mature NBs harbor 11S proteasome components, which are further recruited upon As2O3 exposure. Proteasome recruitment by sumolated PML only likely accounts for the failure of PML-K160R to be degraded. Therefore, studying the basis of As2O3-induced PML/RARalpha degradation we show that PML sumolation directly or indirectly promotes its catabolism, suggesting that mature NBs could be sites of intranuclear proteolysis and opening new insights into NB alterations found in viral infections or transformation.


Adenosine Triphosphatases/metabolism , Arsenicals/pharmacology , Endopeptidases , Neoplasm Proteins/metabolism , Nuclear Matrix/metabolism , Nuclear Proteins , Oxides/pharmacology , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Ubiquitins/metabolism , Amino Acid Motifs , Animals , Arsenic Trioxide , CHO Cells , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Cricetinae , Mice , Models, Biological , Mutation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Promyelocytic Leukemia Protein , Proteasome Endopeptidase Complex , Protein Isoforms/chemistry , Protein Transport , Retinoic Acid Receptor alpha , SUMO-1 Protein , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Suppressor Proteins
...