Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
Haematologica ; 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38841778

IKZF1-deletions occur in 10-15% of patients with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) and predict a poor outcome. However, the impact of IKZF1-loss on sensitivity to drugs used in contemporary treatment protocols has remained underexplored. Here we show in experimental models and in patients that loss of IKZF1 promotes resistance to AraC, a key component of both upfront and relapsed treatment protocols. We attribute this resistance, in part, to diminished import and incorporation of cytarabine (AraC) due to reduced expression of the solute carrier hENT1. Moreover, we find elevated mRNA expression of Evi1, a known driver of therapy resistance in myeloid malignancies. Finally, a kinase directed CRISPR/Cas9-screen identified that inhibition of either mediator kinases CDK8/19 or casein kinase 2 can restore response to AraC. We conclude that this high-risk patient group could benefit from alternative antimetabolites, or targeted therapies that resensitize the cells to AraC.

2.
Pediatr Blood Cancer ; 69(1): e29361, 2022 01.
Article En | MEDLINE | ID: mdl-34597466

INTRODUCTION: One-quarter of the relapses in children with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) occur very early (within 18 months, before completion of treatment), and prognosis in these patients is worse compared to cases that relapse after treatment has ended. METHODS: In this study, we performed a genomic analysis of diagnosis-relapse pairs of 12 children who relapsed very early, followed by a deep-sequencing validation of all identified mutations. In addition, we included one case with a good initial treatment response and on-treatment relapse at the end of upfront therapy. RESULTS: We observed a dynamic clonal evolution in all cases, with relapse almost exclusively originating from a subclone at diagnosis. We identified several driver mutations that may have influenced the outgrowth of a minor clone at diagnosis to become the major clone at relapse. For example, a minimal residual disease (MRD)-based standard-risk patient with ETV6-RUNX1-positive leukemia developed a relapse from a TP53-mutated subclone after loss of the wildtype allele. Furthermore, two patients with TCF3-PBX1-positive leukemia that developed a very early relapse carried E1099K WHSC1 mutations at diagnosis, a hotspot mutation that was recurrently encountered in other very early TCF3-PBX1-positive leukemia relapses as well. In addition to alterations in known relapse drivers, we found two cases with truncating mutations in the cohesin gene RAD21. CONCLUSION: Comprehensive genomic characterization of diagnosis-relapse pairs shows that very early relapses in BCP-ALL frequently arise from minor subclones at diagnosis. A detailed understanding of the therapeutic pressure driving these events may aid the development of improved therapies.


Graft vs Host Disease , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Clonal Evolution/genetics , Genomics , Humans , Prognosis , Recurrence
3.
Cancer Discov ; 11(6): 1322-1323, 2021 06.
Article En | MEDLINE | ID: mdl-34078660

Pikman and colleagues report the results of a multicentric prospective clinical trial of the leukemia precision-based therapy (LEAP) consortium that combines identification of targetable lesions in drug-resistant childhood leukemia, tiered based on evidence for genomic lesions and drug target, validation of matching small-molecule targeted agents, and treatment of individual patients. The study demonstrates the impact of genomic information on disease classification, treatment guidance, and translational research, but also illustrates the challenges for target prediction and trial design for increasingly heterogeneous and smaller subgroups of patients.See related article by Pikman et al., p. 1424.


Antineoplastic Agents , Leukemia , Child , Genomics , Humans , Leukemia/drug therapy , Leukemia/genetics , Prospective Studies , Translational Research, Biomedical
4.
Blood Adv ; 4(19): 4823-4833, 2020 10 13.
Article En | MEDLINE | ID: mdl-33027529

Despite major advances in the treatment of patients with acute lymphoblastic leukemia in the last decades, refractory and/or relapsed disease remains a clinical challenge, and relapsed leukemia patients have an exceedingly dismal prognosis. Dysregulation of apoptotic cell death pathways is a leading cause of drug resistance; thus, alternative cell death mechanisms, such as necroptosis, represent an appealing target for the treatment of high-risk malignancies. We and other investigators have shown that activation of receptor interacting protein kinase 1 (RIP1)-dependent apoptosis and necroptosis by second mitochondria derived activator of caspase mimetics (SMs) is an attractive antileukemic strategy not currently exploited by standard chemotherapy. However, the underlying molecular mechanisms that determine sensitivity to SMs have remained elusive. We show that tumor necrosis factor receptor 2 (TNFR2) messenger RNA expression correlates with sensitivity to SMs in primary human leukemia. Functional genetic experiments using clustered regularly interspaced short palindromic repeats/Cas9 demonstrate that TNFR2 and TNFR1, but not the ligand TNF-α, are essential for the response to SMs, revealing a ligand-independent interplay between TNFR1 and TNFR2 in the induction of RIP1-dependent cell death. Further potential TNFR ligands, such as lymphotoxins, were not required for SM sensitivity. Instead, TNFR2 promotes the formation of a RIP1/TNFR1-containing death signaling complex that induces RIP1 phosphorylation and RIP1-dependent apoptosis and necroptosis. Our data reveal an alternative paradigm for TNFR2 function in cell death signaling and provide a rationale to develop strategies for the identification of leukemias with vulnerability to RIP1-dependent cell death for tailored therapeutic interventions.


Leukemia , Receptors, Tumor Necrosis Factor, Type II , Apoptosis , Caspases , Humans , Leukemia/drug therapy , Leukemia/genetics , Necrosis , Nuclear Pore Complex Proteins , RNA-Binding Proteins , Receptors, Tumor Necrosis Factor, Type II/genetics
5.
Blood Cancer J ; 10(6): 72, 2020 06 26.
Article En | MEDLINE | ID: mdl-32591499

Despite rapid progress in genomic profiling in acute lymphoblastic leukemia (ALL), identification of actionable targets and prediction of response to drugs remains challenging. To identify specific vulnerabilities in ALL, we performed a drug screen using primary human ALL samples cultured in a model of the bone marrow microenvironment combined with high content image analysis. Among the 2487 FDA-approved compounds tested, anthelmintic agents of the class of macrocyclic lactones exhibited potent anti-leukemia activity, similar to the already known anti-leukemia agents currently used in induction chemotherapy. Ex vivo validation in 55 primary ALL samples of both precursor B cell and T-ALL including refractory relapse cases confirmed strong anti-leukemia activity with IC50 values in the low micromolar range. Anthelmintic agents increased intracellular chloride levels in primary leukemia cells, inducing mitochondrial outer membrane depolarization and cell death. Supporting the notion that simultaneously targeting cell death machineries at different angles may enhance the cell death response, combination of anthelmintic agents with the BCL-2 antagonist navitoclax or with the chemotherapeutic agent dexamethasone showed synergistic activity in primary ALL. These data reveal anti-leukemia activity of anthelmintic agents and support exploiting drug repurposing strategies to identify so far unrecognized anti-cancer agents with potential to eradicate even refractory leukemia.


Anthelmintics/pharmacology , Antineoplastic Agents/pharmacology , Drug Repositioning , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Anthelmintics/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Drug Resistance, Neoplasm , Humans , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured , Tumor Microenvironment/drug effects
6.
Clin Cancer Res ; 26(13): 3307-3318, 2020 07 01.
Article En | MEDLINE | ID: mdl-32220889

PURPOSE: Children with Down syndrome (constitutive trisomy 21) that develop acute lymphoblastic leukemia (DS-ALL) have a 3-fold increased likelihood of treatment-related mortality coupled with a higher cumulative incidence of relapse, compared with other children with B-cell acute lymphoblastic leukemia (B-ALL). This highlights the lack of suitable treatment for Down syndrome children with B-ALL. EXPERIMENTAL DESIGN: To facilitate the translation of new therapeutic agents into clinical trials, we built the first preclinical cohort of patient-derived xenograft (PDX) models of DS-ALL, comprehensively characterized at the genetic and transcriptomic levels, and have proven its suitability for preclinical studies by assessing the efficacy of drug combination between the MEK inhibitor trametinib and conventional chemotherapy agents. RESULTS: Whole-exome and RNA-sequencing experiments revealed a high incidence of somatic alterations leading to RAS/MAPK pathway activation in our cohort of DS-ALL, as well as in other pediatric B-ALL presenting somatic gain of the chromosome 21 (B-ALL+21). In murine and human B-cell precursors, activated KRASG12D functionally cooperates with trisomy 21 to deregulate transcriptional networks that promote increased proliferation and self renewal, as well as B-cell differentiation blockade. Moreover, we revealed that inhibition of RAS/MAPK pathway activation using the MEK1/2 inhibitor trametinib decreased leukemia burden in several PDX models of B-ALL+21, and enhanced survival of DS-ALL PDX in combination with conventional chemotherapy agents such as vincristine. CONCLUSIONS: Altogether, using novel and suitable PDX models, this study indicates that RAS/MAPK pathway inhibition represents a promising strategy to improve the outcome of Down syndrome children with B-cell precursor leukemia.


Down Syndrome/complications , Down Syndrome/genetics , Down Syndrome/metabolism , Leukemia, B-Cell/diagnosis , Leukemia, B-Cell/etiology , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Computational Biology/methods , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Humans , Immunophenotyping , Leukemia, B-Cell/therapy , Mice , Mice, Transgenic , Oncogenes , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Signal Transduction/drug effects
7.
Sci Adv ; 5(7): eaau9433, 2019 07.
Article En | MEDLINE | ID: mdl-31392262

Apoptosis is a complex multi-step process driven by caspase-dependent proteolytic cleavage cascades. Dysregulation of apoptosis promotes tumorigenesis and limits the efficacy of chemotherapy. To assess the complex interactions among caspases during apoptosis, we disrupted caspase-8, -9, -3, -7, or -6 and combinations thereof, using CRISPR-based genome editing in living human leukemia cells. While loss of apical initiator caspase-8 or -9 partially blocked extrinsic or intrinsic apoptosis, respectively, only combined loss of caspase-3 and -7 fully inhibited both apoptotic pathways, with no discernible effect of caspase-6 deficiency alone or in combination. Caspase-3/7 double knockout cells exhibited almost complete inhibition of caspase-8 or -9 activation. Furthermore, deletion of caspase-3 and -7 decreased mitochondrial depolarization and cytochrome c release upon apoptosis activation. Thus, activation of effector caspase-3 or -7 sets off explosive feedback amplification of upstream apoptotic events, which is a key feature of apoptotic signaling essential for efficient apoptotic cell death.


Apoptosis/genetics , Caspase 3/genetics , Caspase 7/genetics , Feedback, Physiological , Caspase 8/genetics , Caspase 9/genetics , Cell Polarity/genetics , Cytochromes c/genetics , Gene Knockout Techniques , Humans , Mitochondria/genetics , Signal Transduction/genetics
8.
Front Cell Dev Biol ; 7: 40, 2019.
Article En | MEDLINE | ID: mdl-30941349

Escape from chemotherapy-induced apoptosis is a hallmark of drug resistance in cancer. The recent identification of alternative programmed cell death pathways opens up for possibilities to circumvent the apoptotic blockade in drug resistant cancer and eliminate malignant cells. Indeed, we have recently shown that programmed necrosis, termed necroptosis, could be triggered to induce cell death in a subgroup of primary acute lymphoblastic leukemia (ALL) including highly refractory relapsed cases. In this review we focus on molecular mechanisms that drive drug resistance in ALL of childhood and discuss the potential of necroptosis activation to eradicate resistant disease.

9.
Cancer Cell ; 34(2): 271-285.e7, 2018 08 13.
Article En | MEDLINE | ID: mdl-30107177

The NUP214-ABL1 fusion is a constitutively activated tyrosine kinase that is significantly associated with overexpression of the TLX1 and TLX3 transcription factors in T cell acute lymphoblastic leukemia (T-ALL). Here we show that NUP214-ABL1 cooperates with TLX1 in driving T-ALL development using a transgenic mouse model and human T-ALL cells. Using integrated ChIP-sequencing, ATAC-sequencing, and RNA-sequencing data, we demonstrate that TLX1 and STAT5, the downstream effector of NUP214-ABL1, co-bind poised enhancer regions, and cooperatively activate the expression of key proto-oncogenes such as MYC and BCL2. Inhibition of STAT5, downregulation of TLX1 or MYC, or interference with enhancer function through BET-inhibitor treatment leads to reduction of target gene expression and induction of leukemia cell death.


Enhancer Elements, Genetic , Homeodomain Proteins/physiology , Nuclear Pore Complex Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins/physiology , STAT5 Transcription Factor/physiology , Animals , Gene Fusion , Homeodomain Proteins/genetics , Humans , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/physiology , STAT5 Transcription Factor/genetics
10.
Blood ; 130(3): 297-309, 2017 07 20.
Article En | MEDLINE | ID: mdl-28495792

The interaction of the tumor necrosis factor receptor (TNFR) CD27 with its ligand CD70 is an emerging target to treat cancer. CD27 signaling provides costimulatory signals to cytotoxic T cells but also increases the frequency of regulatory T cells. Similar to other TNFR ligands, CD70 has been shown to initiate intracellular signaling pathways (CD70 reverse signaling). CD27 is expressed on a majority of B-cell non-Hodgkin lymphoma, but its role in the immune control of lymphoma and leukemia is unknown. We therefore generated a cytoplasmic deletion mutant of CD27 (CD27-trunc) to study the role of CD70 reverse signaling in the immunosurveillance of B-cell malignancies in vivo. Expression of CD27-trunc on malignant cells increased the number of tumor-infiltrating interferon γ-producing natural killer (NK) cells. In contrast, the antitumoral T-cell response remained largely unchanged. CD70 reverse signaling in NK cells was mediated via the AKT signaling pathway and increased NK cell survival and effector function. The improved immune control by activated NK cells prolonged survival of CD27-trunc-expressing lymphoma-bearing mice. Finally, CD70 reverse signaling enhanced survival and effector function of human NK cells in a B-cell acute lymphoblastic leukemia xenotransplants model. Therefore, CD70 reverse signaling in NK cells contributes to the immune control of CD27-expressing B-cell lymphoma and leukemia.


B-Lymphocytes/immunology , CD27 Ligand/immunology , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , B-Lymphocytes/pathology , CD27 Ligand/genetics , Gene Expression , Humans , Immunologic Surveillance , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/pathology , Ligands , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction , Survival Analysis , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transplantation, Heterologous , Tumor Necrosis Factor Receptor Superfamily, Member 7/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
11.
Blood ; 129(11): e26-e37, 2017 03 16.
Article En | MEDLINE | ID: mdl-28122742

Drug sensitivity and resistance testing on diagnostic leukemia samples should provide important functional information to guide actionable target and biomarker discovery. We provide proof of concept data by profiling 60 drugs on 68 acute lymphoblastic leukemia (ALL) samples mostly from resistant disease in cocultures of bone marrow stromal cells. Patient-derived xenografts retained the original pattern of mutations found in the matched patient material. Stromal coculture did not prevent leukemia cell cycle activity, but a specific sensitivity profile to cell cycle-related drugs identified samples with higher cell proliferation both in vitro and in vivo as leukemia xenografts. In patients with refractory relapses, individual patterns of marked drug resistance and exceptional responses to new agents of immediate clinical relevance were detected. The BCL2-inhibitor venetoclax was highly active below 10 nM in B-cell precursor ALL (BCP-ALL) subsets, including MLL-AF4 and TCF3-HLF ALL, and in some T-cell ALLs (T-ALLs), predicting in vivo activity as a single agent and in combination with dexamethasone and vincristine. Unexpected sensitivity to dasatinib with half maximal inhibitory concentration values below 20 nM was detected in 2 independent T-ALL cohorts, which correlated with similar cytotoxic activity of the SRC inhibitor KX2-391 and inhibition of SRC phosphorylation. A patient with refractory T-ALL was treated with dasatinib on the basis of drug profiling information and achieved a 5-month remission. Thus, drug profiling captures disease-relevant features and unexpected sensitivity to relevant drugs, which warrants further exploration of this functional assay in the context of clinical trials to develop drug repurposing strategies for patients with urgent medical needs.


Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cells, Cultured , Coculture Techniques , Heterografts , Humans , Mesenchymal Stem Cells/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
12.
Bio Protoc ; 7(7): e2222, 2017 Apr 05.
Article En | MEDLINE | ID: mdl-34541223

CRISPR-Cas9 based knockout strategies are increasingly used to analyze gene function. However, redundancies and overlapping functions in biological signaling pathways can call for generating multi-gene knockout cells, which remains a relatively laborious process. Here we detail the application of multi-color LentiCRISPR vectors to simultaneously generate single and multiple knockouts in human cells. We provide a complete protocol, including guide RNA design, LentiCRISPR cloning, viral production and transduction, as well as strategies for sorting and screening knockout cells. The validity of the process is demonstrated by the simultaneous deletion of up to four programmed cell death mediators in leukemic cell lines and patient-derived acute lymphoblastic leukemia xenografts, in which single cell cloning is not feasible. This protocol enables any lab with access to basic cellular biology equipment, a biosafety level 2 facility and fluorescence-activated cell sorting capabilities to generate single and multi-gene knockout cell lines or primary cells efficiently within one month.

13.
Sci Transl Med ; 8(339): 339ra70, 2016 05 18.
Article En | MEDLINE | ID: mdl-27194728

More precise treatment strategies are urgently needed to decrease toxicity and improve outcomes for treatment-refractory leukemia. We used ex vivo drug response profiling of high-risk, relapsed, or refractory acute lymphoblastic leukemia (ALL) cases and identified a subset with exquisite sensitivity to small-molecule mimetics of the second mitochondria-derived activator of caspases (SMAC) protein. Potent ex vivo activity of the SMAC mimetic (SM) birinapant correlated with marked in vivo antileukemic effects, as indicated by delayed engraftment, decreased leukemia burden, and prolonged survival of xenografted mice. Antileukemic activity was dependent on simultaneous execution of apoptosis and necroptosis, as demonstrated by functional genomic dissection with a multicolored lentiCRISPR approach to simultaneously disrupt multiple genes in patient-derived ALL. SM specifically targeted receptor-interacting protein kinase 1 (RIP1)-dependent death, and CRISPR-mediated disruption of RIP1 completely blocked SM-induced death yet had no impact on the response to standard antileukemic agents. Thus, SM compounds such as birinapant circumvent escape from apoptosis in leukemia by activating a potent dual RIP1-dependent apoptotic and necroptotic cell death, which is not exploited by current therapy. Ex vivo drug activity profiling could provide important functional diagnostic information to identify patients who may benefit from targeted treatment with birinapant in early clinical trials.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Dipeptides/pharmacology , Indoles/pharmacology , Necrosis/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Line, Tumor , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mitochondrial Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
14.
Oncotarget ; 5(22): 11501-12, 2014 Nov 30.
Article En | MEDLINE | ID: mdl-25415224

Interactions with the bone marrow microenvironment are essential for leukemia survival and disease progression. We developed an imaging-based RNAi platform to identify protective cues from bone marrow derived mesenchymal stromal cells (MSC) that promote survival of primary acute lymphoblastic leukemia (ALL) cells. Using a candidate gene approach, we detected distinct responses of individual ALL cases to RNA interference with stromal targets. The strongest effects were observed when interfering with solute carrier family 3 member 2 (SLC3A2) expression, which forms the cystine transporter xc- when associated with SLC7A11. Import of cystine and metabolism to cysteine by stromal cells provides the limiting substrate to generate and maintain glutathione in ALL. This metabolic interaction reduces oxidative stress in ALL cells that depend on stromal xc-. Indeed, cysteine depletion using cysteine dioxygenase resulted in leukemia cell death. Thus, functional evaluation of intercellular interactions between leukemia cells and their microenvironment identifies a selective dependency of ALL cells on stromal metabolism for a relevant subgroup of cases, providing new opportunities to develop more personalized approaches to leukemia treatment.


Cysteine/metabolism , Cytokines/metabolism , Gene Expression Regulation, Leukemic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA Interference , Amino Acid Transport System y+/metabolism , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Cell Death , Cell Survival , Coculture Techniques , Disease Progression , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Humans , Leukemia/metabolism , Mesenchymal Stem Cells/cytology , Oxidative Stress , Reactive Oxygen Species/metabolism , Stromal Cells/metabolism
15.
Int J Cancer ; 133(10): 2341-50, 2013 Nov 15.
Article En | MEDLINE | ID: mdl-23640782

Lymphoproliferative diseases (LPDs) associated with Epstein-Barr virus (EBV) infection cause significant morbidity and mortality in bone marrow and solid organ transplant recipients. To gain insight into LPD pathogenesis and to identify potential effective therapeutic approaches, we investigated early molecular events leading to B-cell transformation by gene expression profiling of EBV-infected B-cells from tonsils by Affymetrix microarray 72 hr postinfection when the B-cells hyperproliferation phase starts. Cell cycle and apoptosis were the most significantly affected pathways and enriched gene sets. In particular, we found significantly increased expression of cyclin-dependent kinase (CDK)1 and CCNB1 (cyclin B1) and of one of their downstream targets BIRC5 (survivin). Importantly, the strong upregulation of the antiapoptotic protein survivin was confirmed in lymphoblastoid cell lines (LCLs) and 71% of EBV-positive post-transplant EBV-LPD lesions scored positive for survivin. The validity of early transforming events for the identification of therapeutic targets for EBV-LPD was confirmed by the marked antiproliferative effect of the CDK inhibitor flavopiridol on LCLs and by the strong induction of apoptosis by survivin inhibition with YM155 or terameprocol. Our results suggest that targeting of CDKs and/or survivin in post-transplant EBV-LPD by specific inhibitors might be an important approach to control and eliminate EBV-transformed B-cells that should be further considered.


B-Lymphocytes/metabolism , B-Lymphocytes/virology , CDC2 Protein Kinase/genetics , Epstein-Barr Virus Infections/genetics , Inhibitor of Apoptosis Proteins/genetics , Lymphoproliferative Disorders/genetics , Organ Transplantation/adverse effects , Apoptosis/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle/genetics , Cyclin B1/genetics , Cyclin B1/metabolism , Epstein-Barr Virus Infections/etiology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Gene Expression , Herpesvirus 4, Human , Humans , Inhibitor of Apoptosis Proteins/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/metabolism , Survivin , Transformation, Genetic , Up-Regulation
16.
Blood ; 121(25): e149-59, 2013 Jun 20.
Article En | MEDLINE | ID: mdl-23649467

A better description of the leukemia cell surface proteome (surfaceome) is a prerequisite for the development of diagnostic and therapeutic tools. Insights into the complexity of the surfaceome have been limited by the lack of suitable methodologies. We combined a leukemia xenograft model with the discovery-driven chemoproteomic Cell Surface Capture technology to explore the B-cell precursor acute lymphoblastic leukemia (BCP-ALL) surfaceome; 713 cell surface proteins, including 181 CD proteins, were detected through combined analysis of 19 BCP-ALL cases. Diagnostic immunophenotypes were recapitulated in each case, and subtype specific markers were detected. To identify new leukemia-associated markers, we filtered the surfaceome data set against gene expression information from sorted, normal hematopoietic cells. Nine candidate markers (CD18, CD63, CD31, CD97, CD102, CD157, CD217, CD305, and CD317) were validated by flow cytometry in patient samples at diagnosis and during chemotherapy. CD97, CD157, CD63, and CD305 accounted for the most informative differences between normal and malignant cells. The ALL surfaceome constitutes a valuable resource to assist the functional exploration of surface markers in normal and malignant lymphopoiesis. This unbiased approach will also contribute to the development of strategies that rely on complex information for multidimensional flow cytometry data analysis to improve its diagnostic applications.


Biomarkers, Tumor/analysis , Membrane Proteins/analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proteome/analysis , Animals , Antigens, CD/analysis , Flow Cytometry , Humans , Immunophenotyping , Membrane Proteins/metabolism , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Proteome/metabolism , Xenograft Model Antitumor Assays
17.
J Biol Chem ; 287(16): 12602-11, 2012 Apr 13.
Article En | MEDLINE | ID: mdl-22378787

The LDLR is a critical factor in the regulation of blood cholesterol levels that are altered in different human diseases. The level of LDLR in the cell is regulated by both transcriptional and post-transcriptional events. The E3 ubiquitin ligase, myosin regulatory light chain-interacting protein (Mylip)/inducible degrader of the LDL-R (Idol) was shown to induce degradation of LDLR via protein ubiquitination. We have here studied novel factors and mechanisms that may regulate Mylip/Idol in human hepatocyte cells and in mouse macrophages. We observed that FGF21 that is present in serum in different conditions reduced Mylip/Idol at the RNA and protein level, and increased LDLR levels and stability in the cells. FGF21 also enhanced expression of Canopy2 (Cnpy2)/MIR-interacting Saposin-like protein (Msap) that is known to interact with Mylip/Idol. Overexpression of Cnpy2/Msap increased LDLRs, and knockdown experiments showed that Cnpy2/Msap is crucial for the FGF21 effect on LDLRs. Experiments using DiI-labeled LDL particles showed that FGF21 increased lipoprotein uptake and the effect of FGF21 was additive to that of statins. Our results are consistent with an important role of FGF21 and Cnpy2/Msap in the regulation of LDLRs in cultured cells, which warrants further studies using human samples.


Adaptor Proteins, Signal Transducing/metabolism , Cholesterol/pharmacokinetics , Fibroblast Growth Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, LDL/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Membrane Proteins/genetics , Mice , Ubiquitin-Protein Ligases/genetics , Ubiquitination/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
19.
Blood ; 118(7): 1854-64, 2011 Aug 18.
Article En | MEDLINE | ID: mdl-21670474

Clonal evolution of the leukemogenic compartment may contribute to alter the therapeutic response in acute lymphoblastic leukemia (ALL). Using xenotransplantation of primary leukemia cells, we evaluated the phenotypic and genetic composition of de novo resistant very high risk precursor B-cell ALL, a subgroup defined by the persistence of minimal residual disease despite intensive chemotherapy. Analysis of copy number alterations (CNAs) showed that the xenografted leukemia, even when reconstituted from 100 cells, remained highly related to the diagnostic sample, with minor changes in CNAs, mostly deletions, emerging in most cases in the first passage into mice. At the single-cell level, the pattern of monoallelic and biallelic deletions of the CDKN2A locus revealed distinct leukemia subpopulations, which were reproducibly tracked in xenografts. In most very high risk ALL cases, the predominant diagnostic clones were reconstituted in xenografts, as shown by multiplex polymerase chain reaction analysis of immunoglobulin and T-cell receptor loci. In other cases, the pattern in CNAs and immunoglobulin and T-cell receptor rearrangement was less concordant in xenografts, suggesting the outgrowth of subclones. These results unequivocally demonstrate the existence of clonally closely related but distinct subsets of leukemia initiating cells in ALL, which has important implications for drug development and preclinical disease modeling.


Phenotype , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Antineoplastic Agents/therapeutic use , Clone Cells , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Deletion , Gene Dosage , Gene Rearrangement , Gene Rearrangement, T-Lymphocyte , Genes, Immunoglobulin , Humans , Mice , Neoplasm, Residual , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Transplantation, Heterologous , Tumor Cells, Cultured
20.
PLoS One ; 6(5): e19506, 2011.
Article En | MEDLINE | ID: mdl-21602936

Cancer stem cells (CSCs) have been identified in a number of solid tumors, but not yet in rhabdomyosarcoma (RMS), the most frequently occurring soft tissue tumor in childhood. Hence, the aim of this study was to identify and characterize a CSC population in RMS using a functional approach. We found that embryonal rhabdomyosarcoma (eRMS) cell lines can form rhabdomyosarcoma spheres (short rhabdospheres) in stem cell medium containing defined growth factors over several passages. Using an orthotopic xenograft model, we demonstrate that a 100 fold less sphere cells result in faster tumor growth compared to the adherent population suggesting that CSCs were enriched in the sphere population. Furthermore, stem cell genes such as oct4, nanog, c-myc, pax3 and sox2 are significantly upregulated in rhabdospheres which can be differentiated into multiple lineages such as adipocytes, myocytes and neuronal cells. Surprisingly, gene expression profiles indicate that rhabdospheres show more similarities with neuronal than with hematopoietic or mesenchymal stem cells. Analysis of these profiles identified the known CSC marker CD133 as one of the genes upregulated in rhabdospheres, both on RNA and protein levels. CD133(+) sorted cells were subsequently shown to be more tumorigenic and more resistant to commonly used chemotherapeutics. Using a tissue microarray (TMA) of eRMS patients, we found that high expression of CD133 correlates with poor overall survival. Hence, CD133 could be a prognostic marker for eRMS. These experiments indicate that a CD133(+) CSC population can be enriched from eRMS which might help to develop novel targeted therapies against this pediatric tumor.


Antigens, CD/analysis , Glycoproteins/analysis , Neoplastic Stem Cells/pathology , Peptides/analysis , Rhabdomyosarcoma, Embryonal/pathology , AC133 Antigen , Animals , Antigens, CD/genetics , Biomarkers, Tumor , Cell Culture Techniques , Cell Differentiation , Cell Line, Tumor , Gene Expression Profiling , Glycoproteins/genetics , Humans , Mice , Peptides/genetics , Prognosis , Rhabdomyosarcoma, Embryonal/diagnosis , Survival Rate , Tissue Array Analysis , Transplantation, Heterologous
...