91 results on '"Stam RW"'
Search Results
2. Src kinase-induced phosphorylation of annexin A2 mediates glucocorticoid resistance in MLL-rearranged infant acute lymphoblastic leukemia
- Author
-
Spijkers-Hagelstein, JAP, Mimoso Pinhanços, S, Schneider, P, Pieters, R, and Stam, RW
- Published
- 2013
- Full Text
- View/download PDF
3. A phase 1/2, open-label, dose-escalation study of midostaurin in children with relapsed or refractory acute leukaemia
- Author
-
Zwaan, C, Söderhäll, S, Brethon, B, Luciani, M, Rizzari, C, Stam, R, Besse, E, Dutreix, C, Fagioli, F, Ho, P, Dufour, C, Pieters, R, Zwaan, CM, Stam, RW, Ho, PA, Zwaan, C, Söderhäll, S, Brethon, B, Luciani, M, Rizzari, C, Stam, R, Besse, E, Dutreix, C, Fagioli, F, Ho, P, Dufour, C, Pieters, R, Zwaan, CM, Stam, RW, and Ho, PA
- Published
- 2019
4. Revisiting the biology of infant t(4;11)/MLL-AF4(+) B-cell acute lymphoblastic leukemia
- Author
-
Sanjuan-Pla A, Bueno C, Prieto C, Acha P, Stam RW, Marschalek R, and Menéndez P
- Subjects
hemic and lymphatic diseases - Abstract
Infant B-cell acute lymphoblastic leukemia (B-ALL) accounts for 10% of childhood ALL. The genetic hallmark of most infant B-ALL is chromosomal rearrangements of the mixed-lineage leukemia (MLL) gene. Despite improvement in the clinical management and survival (similar to 85-90%) of childhood B-ALL, the outcome of infants with MLL-rearranged (MLL-r) B-ALL remains dismal, with overall survival
- Published
- 2015
5. Prognostic significance of high-level FLT3 expression in MLL-rearranged infant acute lymphoblastic leukemia [8]
- Author
-
Stam, RW, Schneider, P, de Lorenzo, P, den Boer, ML, Pieters, R., VALSECCHI, MARIA GRAZIA, Stam, R, Schneider, P, de Lorenzo, P, Valsecchi, M, den Boer, M, and Pieters, R
- Subjects
Gene Expression Regulation, Neoplastic ,fms-Like Tyrosine Kinase 3 ,Prognosi ,Infant ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Myeloid-Lymphoid Leukemia Protein - Abstract
Recently, we have demonstrated that in primary MLLrearranged infant acute lymphoblastic leukemia (ALL) samples, high-level expression of wild-type FLT3 is associated with FLT3 phosphorylation (ie, constitutive activation) and sensitivity toward the small-molecule FLT3 inhibitor PKC412.1,2 Similarly, Brown et al reported that MLL-rearranged ALL cells with high-level FLT3 expression are selectively killed by the FLT3 inhibitor CEP-701.3 These findings suggest the existence of a threshold level of FLT3 expression above which activated FLT3 becomes apparent and “targetable.” Exploring this possibility, we have analyzed the prognosis of our recently published cohort of patients with MLL-rearranged infantALL2 in relation to FLT3 expression. For 32 of 41 samples for which the level of FLT3 expression was determined using quantitative RT-PCR, clinical data were available. All of these samples tested negative for the presence of activating FLT3/tyrosine kinase domain (TKD) mutations or FLT3/ internal tandem duplications (ITDs). Among these 32 patients with MLL-rearranged infant ALL, FLT3 expression relative to the housekeeping gene GAPDH ranged from 0.46% to 17.4%, with a median and mean expression level of 1.8% and 3.5%, respectively. Moreover, all patients were uniformly treated according the Interfant-99 protocol. Using the mean FLT3 expression as the cut-off value to divide patients into 2 groups expressing low or high levels of FLT3, we found that high-level FLT3 expression is likely associated with a poor treatment outcome within this subtype of ALL that is already characterized by a poor prognosis.4–6 The 1-year event-free survival (EFS) estimate is 71% for patients expressing low levels of FLT3, compared with 36% for patients displaying high-level FLT3 expression. These data are in concordance with data published by Ozeki et al,7 who showed that high-level expression of wild-type FLT3 is an unfavorable prognostic factor for overall survival in acute myeloid leukemia (AML). We conclude that constitutively activated FLT3 as a result of increased expression may contribute to the dismal prognosis of MLL-rearranged infant ALL. However, this should be confirmed in a larger prospective study. Targeting FLT3 overexpression in MLL-rearranged infantALL using the FLT3 inhibitors PKC412 and CEP-701 has been shown to be effective in vitro and in vivo in a mouse model.1–3 Based upon these findings, The Childhood Oncology Group in the United States, and the Interfant Study Group worldwide are planning clinical studies including these FLT3 inhibitors. The finding we present here emphasizes the value of these clinical trials, as we show that the patients that are most likely to respond to FLT3 inhibitors are patients at very high risk of treatment failure.
- Published
- 2007
6. Frequencies and prognostic impact of RAS mutations in MLL-rearranged acute lymphoblastic leukemia in infants
- Author
-
Driessen, E, van Roon, E, Spijkers Hagelstein, J, Schneider, P, de Lorenzo, P, Valsecchi, M, Pieters, R, Stam, R, Driessen, EM, van Roon, EH, Spijkers Hagelstein, JA, VALSECCHI, MARIA GRAZIA, Stam, RW, Driessen, E, van Roon, E, Spijkers Hagelstein, J, Schneider, P, de Lorenzo, P, Valsecchi, M, Pieters, R, Stam, R, Driessen, EM, van Roon, EH, Spijkers Hagelstein, JA, VALSECCHI, MARIA GRAZIA, and Stam, RW
- Abstract
Acute lymphoblastic leukemia in infants represents an aggressive malignancy associated with a high incidence (approx. 80%) of translocations involving the Mixed Lineage Leukemia (MLL) gene. Attempts to mimic Mixed Lineage Leukemia fusion driven leukemogenesis in mice raised the question whether these fusion proteins require secondary hits. RAS mutations are suggested as candidates. Earlier results on the incidence of RAS mutations in Mixed Lineage Leukemia-rearranged acute lymphoblastic leukemia are inconclusive. Therefore, we studied frequencies and relation with clinical parameters of RAS mutations in a large cohort of infant acute lymphoblastic leukemia patients. Using conventional sequencing analysis, we screened neuroblastoma RAS viral (v-ras) oncogene homolog gene (NRAS), v-Ki-ras Kirsten rat sarcoma viral oncogene homolog gene (KRAS), and v-raf murine sarcoma viral oncogene homolog B1 gene (BRAF) for mutations in a large cohort (n=109) of infant acute lymphoblastic leukemia patients and studied the mutations in relation to several clinical parameters, and in relation to Homeobox gene A9 expression and the presence of ALL1 fused gene 4-Mixed Lineage Leukemia (AF4-MLL). Mutations were detected in approximately 14% of all cases, with a higher frequency of approximately 24% in t(4;11)-positive patients (P=0.04). Furthermore, we identified RAS mutations as an independent predictor (P=0.019) for poor outcome in Mixed Lineage Leukemia-rearranged infant acute lymphoblastic leukemia, with a hazard ratio of 3.194 (95% confidence interval (CI):1.211-8.429). Also, RAS-mutated infants have higher white blood cell counts at diagnosis (P=0.013), and are more resistant to glucocorticoids in vitro (P<0.05). Finally, we demonstrate that RAS mutations, and not the lack of Homeobox gene A9 expression nor the expression of AF4-MLL are associated with poor outcome in t(4;11)-rearranged infants. We conclude that the presence of RAS mutations in Mixed Lineage Leukemia-rearranged
- Published
- 2013
7. Gene expression profiling–based dissection of MLL translocated and MLL germline acute lymphoblastic leukemia in infants
- Author
-
Stam, R, Schneider, P, Hagelstein, J, van der Linden, M, Stumpel, D, de Menezes, R, de Lorenzo, P, Valsecchi, M, Pieters, R, Stam, RW, Hagelstein, JA, van der Linden, MH, Stumpel, DJ, de Menezes, RX, Pieters, R., VALSECCHI, MARIA GRAZIA, Stam, R, Schneider, P, Hagelstein, J, van der Linden, M, Stumpel, D, de Menezes, R, de Lorenzo, P, Valsecchi, M, Pieters, R, Stam, RW, Hagelstein, JA, van der Linden, MH, Stumpel, DJ, de Menezes, RX, Pieters, R., and VALSECCHI, MARIA GRAZIA
- Abstract
Acute lymphoblastic leukemia (ALL) in infants (< 1 year) is characterized by a poor prognosis and a high incidence of MLL translocations. Several studies demonstrated the unique gene expression profile associated with MLL-rearranged ALL, but generally small cohorts were analyzed as uniform patient groups regardless of the type of MLL translocation, whereas the analysis of translocationnegative infant ALL remained unacknowledged. Here we generated and analyzed primary infant ALL expression profiles (n 73) typified by translocations t(4;11), t(11;19), and t(9;11), or the absence of MLL translocations. Our data show that MLL germline infant ALL specifies a gene expression pattern that is different from both MLL-rearranged infantALL and pediatric precursor B-ALL. Moreover, we demonstrate that, apart from a fundamental signature shared by all MLL-rearranged infant ALL samples, each type of MLL translocation is associated with a translocation- specific gene expression signature. Finally, we show the existence of 2 distinct subgroups among t(4;11)– positive infant ALL cases characterized by the absence or presence of HOXA expression, and that patients lacking HOXA expression are at extreme high risk of disease relapse. These gene expression profiles should provide important novel insights in the complex biology of MLL-rearranged infant ALL and boost our progress in finding novel therapeutic solutions.
- Published
- 2010
8. Association of high-level MCL-1 expression with in vitro and in vivo prednisone resistance in MLL-rearranged infant acute lymphoblastic leukemia
- Author
-
Stam, R, Den Boer, M, Schneider, P, de Boer, J, Hagelstein, J, Valsecchi, M, de Lorenzo, P, Sallan, S, Brady, H, Armstrong, S, Pieters, R, Stam, RW, Den Boer, ML, Sallan, SE, Brady, HJ, Armstrong, SA, Pieters, R., VALSECCHI, MARIA GRAZIA, Stam, R, Den Boer, M, Schneider, P, de Boer, J, Hagelstein, J, Valsecchi, M, de Lorenzo, P, Sallan, S, Brady, H, Armstrong, S, Pieters, R, Stam, RW, Den Boer, ML, Sallan, SE, Brady, HJ, Armstrong, SA, Pieters, R., and VALSECCHI, MARIA GRAZIA
- Abstract
MLL-rearranged acute lymphoblastic leukemia (ALL) represents an unfavorable type of leukemia that often is highly resistant to glucocorticoids such as prednisone and dexamethasone. Because response to prednisone largely determines clinical outcome of pediatric patients with ALL, overcoming resistance to this drug may be an important step toward improving prognosis. Here, we show how gene expression profiling identifies high-level MCL-1 expression to be associated with prednisolone resistance in MLL-rearranged infant ALL, as well as in more favorable types of childhood ALL. To validate this observation, we determined MCL-1 expression with quantitative reverse transcription–polymerase chain reaction in a cohort of MLL-rearranged infant ALL and pediatric noninfant ALL samples and confirmed that high-level MCL-1 expression is associated with prednisolone resistance in vitro. In addition, MCL-1 expression appeared to be significantly higher in MLL-rearranged infant patients who showed a poor response to prednisone in vivo compared with prednisone good responders. Finally, downregulation of MCL-1 in prednisoloneresistant MLL-rearranged leukemia cells by RNA interference, to some extent, led to prednisolone sensitization. Collectively, our findings suggest a potential role for MCL-1 in glucocorticoid resistance in MLL-rearranged infant ALL, but at the same time strongly imply that highlevel MCL-1 expression is not the sole mechanism providing resistance to these drugs.
- Published
- 2010
9. Specific promoter methylation identifies different subgroups of MLL-rearranged infant acute lymphoblastic leukemia, influences clinical outcome, and provides therapeutic options
- Author
-
Stumpel, D, Schneider, P, van Roon, E, Boer, J, de Lorenzo, P, Valsecchi, M, de Menezes, R, Pieters, R, Stam, R, Stumpel, DJ, van Roon, EH, Boer, JM, de Menezes, RX, Stam, RW, VALSECCHI, MARIA GRAZIA, Stumpel, D, Schneider, P, van Roon, E, Boer, J, de Lorenzo, P, Valsecchi, M, de Menezes, R, Pieters, R, Stam, R, Stumpel, DJ, van Roon, EH, Boer, JM, de Menezes, RX, Stam, RW, and VALSECCHI, MARIA GRAZIA
- Abstract
MLL-rearranged infant acute lymphoblastic leukemia (ALL) remains the most aggressive type of childhood leukemia, displaying a unique gene expression profile. Here we hypothesized that this characteristic gene expression signature may have been established by potentially reversible epigenetic modifications. To test this hypothesis, we used differential methylation hybridization to explore the DNA methylation patterns underlying MLLrearranged ALL in infants. The obtained results were correlated with gene expression data to confirm gene silencing as a result of promoter hypermethylation. Distinct promoter CpG island methylation patterns separated different genetic subtypes of MLL-rearranged ALL in infants. MLL translocations t(4;11) and t(11;19) characterized extensively hypermethylated leukemias, whereas t(9;11)-positive infant ALL and infant ALL carrying wildtype MLL genes epigenetically resembled normal bone marrow. Furthermore, the degree of promoter hypermethylation among infant ALL patients carrying t(4; 11) or t(11;19) appeared to influence relapse-free survival, with patients displaying accentuated methylation being at high relapse risk. Finally, we show that the demethylating agent zebularine reverses aberrant DNA methylation and effectively induces apoptosis in MLLrearranged ALL cells. Collectively these data suggest that aberrant DNA methylation occurs in the majority of MLLrearranged infant ALL cases and guides clinical outcome. Therefore, inhibition of aberrant DNA methylation may be an important novel therapeutic strategy for MLL-rearranged ALL in infants.
- Published
- 2009
10. Prognostic significance of high-level FLT3 expression in MLL-rearranged infant acute lymphoblastic leukemia [8]
- Author
-
Stam, R, Schneider, P, de Lorenzo, P, Valsecchi, M, den Boer, M, Pieters, R, Stam, RW, den Boer, ML, Pieters, R., VALSECCHI, MARIA GRAZIA, Stam, R, Schneider, P, de Lorenzo, P, Valsecchi, M, den Boer, M, Pieters, R, Stam, RW, den Boer, ML, Pieters, R., and VALSECCHI, MARIA GRAZIA
- Abstract
Recently, we have demonstrated that in primary MLLrearranged infant acute lymphoblastic leukemia (ALL) samples, high-level expression of wild-type FLT3 is associated with FLT3 phosphorylation (ie, constitutive activation) and sensitivity toward the small-molecule FLT3 inhibitor PKC412.1,2 Similarly, Brown et al reported that MLL-rearranged ALL cells with high-level FLT3 expression are selectively killed by the FLT3 inhibitor CEP-701.3 These findings suggest the existence of a threshold level of FLT3 expression above which activated FLT3 becomes apparent and “targetable.” Exploring this possibility, we have analyzed the prognosis of our recently published cohort of patients with MLL-rearranged infantALL2 in relation to FLT3 expression. For 32 of 41 samples for which the level of FLT3 expression was determined using quantitative RT-PCR, clinical data were available. All of these samples tested negative for the presence of activating FLT3/tyrosine kinase domain (TKD) mutations or FLT3/ internal tandem duplications (ITDs). Among these 32 patients with MLL-rearranged infant ALL, FLT3 expression relative to the housekeeping gene GAPDH ranged from 0.46% to 17.4%, with a median and mean expression level of 1.8% and 3.5%, respectively. Moreover, all patients were uniformly treated according the Interfant-99 protocol. Using the mean FLT3 expression as the cut-off value to divide patients into 2 groups expressing low or high levels of FLT3, we found that high-level FLT3 expression is likely associated with a poor treatment outcome within this subtype of ALL that is already characterized by a poor prognosis.4–6 The 1-year event-free survival (EFS) estimate is 71% for patients expressing low levels of FLT3, compared with 36% for patients displaying high-level FLT3 expression. These data are in concordance with data published by Ozeki et al,7 who showed that high-level expression of wild-type FLT3 is an unfavorable prognostic factor for overall survival in acute myeloid leukemia
- Published
- 2007
11. Targeting FLT3 in primary MLL-gene-rearranged infant acute lymphoblastic leukemia
- Author
-
Stam, R, den Boer, M, Schneider, P, Nollau, P, Horstmann, M, Beverloo, H, van der Voort, E, Valsecchi, M, de Lorenzo, P, Sallan, S, Armstrong, S, Pieters, R, Stam, RW, den Boer, ML, Beverloo, HB, Sallan, SE, Armstrong, SA, Pieters, R., VALSECCHI, MARIA GRAZIA, Stam, R, den Boer, M, Schneider, P, Nollau, P, Horstmann, M, Beverloo, H, van der Voort, E, Valsecchi, M, de Lorenzo, P, Sallan, S, Armstrong, S, Pieters, R, Stam, RW, den Boer, ML, Beverloo, HB, Sallan, SE, Armstrong, SA, Pieters, R., and VALSECCHI, MARIA GRAZIA
- Abstract
Acute lymphoblastic leukemia (ALL) in infants is characterized by rearrangements of the mixed lineage leukemia (MLL) gene, drug resistance, and a poor treatment outcome. Therefore, novel therapeutic strategies are needed to improve prognosis. Recently, we showed that FLT3 is highly expressed in MLL rearranged ALL (MLL). Here we demonstrate FLT3 expression in infants with MLL (n = 41) to be significantly higher compared to both infant (n = 8; P < .001) and noninfant patients with ALL (n = 23; P = .001) carrying germline MLL genes. Furthermore, leukemic cells from infants with MLL were significantly more sensitive to the Fms-like tyrosine kinase 3 (FLT3) inhibitor PKC412 (N-benzoyl staurosporine) than noninfant ALL cells, and at least as sensitive as internal tandem duplication-positive (ITD+) AML cells. Surprisingly, activation loop mutations only occurred in about 3% (1 of 36) of the cases and no FLT3/ITDs were observed. However, measuring FLT3 phosphorylation in infants with MLL expressing varying levels of wild-type FLT3 revealed that high-level FLT3 expression is associated with ligand-independent FLT3 activation. This suggests that infant MLL cells displaying activated FLT3 as a result of overexpression can be targeted by FLT3 inhibitors such as PKC412. However, at concentrations of PKC412 minimally required to fully inhibit FLT3 phosphorylation, the cytotoxic effects were only fractional. Thus, PKC412-induced apoptosis in infant MLL cells is unlikely to be a consequence of FLT3 inhibition alone but may involve inhibition of multiple other kinases by this drug.
- Published
- 2005
12. NG2 is a target gene of MLL-AF4 and underlies glucocorticoid resistance in MLLr B-ALL by regulating NR3C1 expression.
- Author
-
Lopez-Millan B, Rubio-Gayarre A, Vinyoles M, Trincado JL, Fraga MF, Fernandez-Fuentes N, Guerrero-Murillo M, Martinez A, Velasco-Hernandez T, Falgàs A, Panisello C, Valcarcel G, Sardina JL, López-Martí P, Javierre BM, Del Valle-Pérez B, García de Herreros A, Locatelli F, Pieters R, Bardini M, Cazzaniga G, Rodríguez-Manzaneque JC, Hanewald T, Marschalek R, Milne TA, Stam RW, Tejedor JR, Menendez P, and Bueno C
- Subjects
- Humans, Gene Expression Regulation, Leukemic, Cell Line, Tumor, Animals, Mice, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Antigens, Proteoglycans, Receptors, Glucocorticoid metabolism, Receptors, Glucocorticoid genetics, Myeloid-Lymphoid Leukemia Protein genetics, Myeloid-Lymphoid Leukemia Protein metabolism, Drug Resistance, Neoplasm genetics, Glucocorticoids pharmacology, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism
- Abstract
Abstract: B-cell acute lymphoblastic leukemia (B-ALL) is the most common pediatric cancer, with long-term overall survival rates of ∼85%. However, B-ALL harboring rearrangements of the MLL gene (also known as KMT2A), referred to as MLLr B-ALL, is common in infants and is associated with poor 5-year survival, relapses, and refractoriness to glucocorticoids (GCs). GCs are an essential part of the treatment backbone for B-ALL, and GC resistance is a major clinical predictor of poor outcome. Elucidating the mechanisms of GC resistance in MLLr B-ALL is, therefore, critical to guide therapeutic strategies that deepen the response after induction therapy. Neuron-glial antigen-2 (NG2) expression is a hallmark of MLLr B-ALL and is minimally expressed in healthy hematopoietic cells. We recently reported that NG2 expression is associated with poor prognosis in MLLr B-ALL. Despite its contribution to MLLr B-ALL pathogenesis, the role of NG2 in MLLr-mediated leukemogenesis/chemoresistance remains elusive. Here, we show that NG2 is an epigenetically regulated direct target gene of the leukemic MLL-ALF transcription elongation factor 4 (AF4) fusion protein. NG2 negatively regulates the expression of the GC receptor nuclear receptor subfamily 3 group C member 1 (NR3C1) and confers GC resistance to MLLr B-ALL cells. Mechanistically, NG2 interacts with FLT3 to render ligand-independent activation of FLT3 signaling (a hallmark of MLLr B-ALL) and downregulation of NR3C1 via activating protein-1 (AP-1)-mediated transrepression. Collectively, our study elucidates the role of NG2 in GC resistance in MLLr B-ALL through FLT3/AP-1-mediated downregulation of NR3C1, providing novel therapeutic avenues for MLLr B-ALL., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
13. Selective inhibition of HDAC class IIA as therapeutic intervention for KMT2A-rearranged acute lymphoblastic leukemia.
- Author
-
Verbeek TCAI, Vrenken KS, Arentsen-Peters STCJM, Castro PG, van de Ven M, van Tellingen O, Pieters R, and Stam RW
- Subjects
- Humans, Animals, Mice, Histone Deacetylases genetics, Histone Deacetylases metabolism, Cell Line, Tumor, Xenograft Model Antitumor Assays, Gene Rearrangement, Apoptosis drug effects, Female, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Myeloid-Lymphoid Leukemia Protein genetics, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylase Inhibitors therapeutic use, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase antagonists & inhibitors
- Abstract
KMT2A-rearranged acute lymphoblastic leukemia (ALL) is characterized by deregulation of the epigenome and shows susceptibility towards histone deacetylase (HDAC) inhibition. Most broad-spectrum HDAC inhibitors simultaneously target multiple human HDAC isoforms. Consequently, they often induce toxicity and especially in combination with other therapeutic agents. Therefore, more specifically targeting HDAC isoforms may represent a safer therapeutic strategy. Here we show that shRNA-mediated knock-down of the class IIA HDAC isoforms HDAC4, HDAC5, and HDAC7 results in apoptosis induction and cell cycle arrest in KMT2A-rearranged ALL cells. In concordance, the HDAC4/5 selective small molecule inhibitor LMK-235 effectively eradicates KMT2A-rearranged ALL cell lines as well as primary patient samples in vitro. However, using a xenograft mouse model of KMT2A-rearranged ALL we found that the maximum achievable dose of LMK-235 was insufficient to induce anti-leukemic effects in vivo. Similar results were obtained for the specific class IIA HDAC inhibitors MC1568 and TMP195. Finally, LMK-235 appeared to exert minimal anti-leukemic effects in vivo in combination with the BCL-2 inhibitor venetoclax, but not enough to prolong survival in treated mice. In conclusion, class IIA HDAC isoforms represent attractive therapeutic target in KMT2A-rearranged ALL, although clinical applications require the development of more stable and efficient specific HDAC inhibitors., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
14. Distinct Responses to Menin Inhibition and Synergy with DOT1L Inhibition in KMT2A -Rearranged Acute Lymphoblastic and Myeloid Leukemia.
- Author
-
Adriaanse FRS, Schneider P, Arentsen-Peters STCJM, Fonseca AMND, Stutterheim J, Pieters R, Zwaan CM, and Stam RW
- Subjects
- Humans, Drug Synergism, Gene Rearrangement, Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Drug Resistance, Neoplasm drug effects, Mutation, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Methyltransferases antagonists & inhibitors, Methyltransferases genetics, Methyltransferases metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology
- Abstract
Pediatric acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) exhibit favorable survival rates. However, for AML and ALL patients carrying KMT2A gene translocations clinical outcome remains unsatisfactory. Key players in KMT2A-fusion-driven leukemogenesis include menin and DOT1L. Recently, menin inhibitors like revumenib have garnered attention for their potential therapeutic efficacy in treating KMT2A -rearranged acute leukemias. However, resistance to menin inhibition poses challenges, and identifying which patients would benefit from revumenib treatment is crucial. Here, we investigated the in vitro response to revumenib in KMT2A -rearranged ALL and AML. While ALL samples show rapid, dose-dependent induction of leukemic cell death, AML responses are much slower and promote myeloid differentiation. Furthermore, we reveal that acquired resistance to revumenib in KMT2A -rearranged ALL cells can occur either through the acquisition of MEN1 mutations or independently of mutations in MEN1 . Finally, we demonstrate significant synergy between revumenib and the DOT1L inhibitor pinometostat in KMT2A -rearranged ALL, suggesting that such drug combinations represent a potent therapeutic strategy for these patients. Collectively, our findings underscore the complexity of resistance mechanisms and advocate for precise patient stratification to optimize the use of menin inhibitors in KMT2A -rearranged acute leukemia.
- Published
- 2024
- Full Text
- View/download PDF
15. Combining CRISPR-Cas9 and TCR exchange to generate a safe and efficient cord blood-derived T cell product for pediatric relapsed AML.
- Author
-
Lo Presti V, Meringa A, Dunnebach E, van Velzen A, Moreira AV, Stam RW, Kotecha RS, Krippner-Heidenreich A, Heidenreich OT, Plantinga M, Cornel A, Sebestyen Z, Kuball J, van Til NP, and Nierkens S
- Subjects
- Humans, Child, CD8-Positive T-Lymphocytes, CRISPR-Cas Systems genetics, Fetal Blood, Receptors, Antigen, T-Cell genetics, Cell Line, Tumor, Recurrence, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Antineoplastic Agents
- Abstract
Background: Hematopoietic cell transplantation (HCT) is an effective treatment for pediatric patients with high-risk, refractory, or relapsed acute myeloid leukemia (AML). However, a large proportion of transplanted patients eventually die due to relapse. To improve overall survival, we propose a combined strategy based on cord blood (CB)-HCT with the application of AML-specific T cell receptor (TCR)-engineered T cell therapy derived from the same CB graft., Methods: We produced CB-CD8
+ T cells expressing a recombinant TCR (rTCR) against Wilms tumor 1 (WT1) while lacking endogenous TCR (eTCR) expression to avoid mispairing and competition. CRISPR-Cas9 multiplexing was used to target the constant region of the endogenous TCRα ( TRAC ) and TCRβ ( TRBC ) chains. Next, an optimized method for lentiviral transduction was used to introduce recombinant WT1-TCR. The cytotoxic and migration capacity of the product was evaluated in coculture assays for both cell lines and primary pediatric AML blasts., Results: The gene editing and transduction procedures achieved high efficiency, with up to 95% of cells lacking eTCR and over 70% of T cells expressing rWT1-TCR. WT1-TCR-engineered T cells lacking the expression of their eTCR (eTCR-/- WT1-TCR) showed increased cell surface expression of the rTCR and production of cytotoxic cytokines, such as granzyme A and B, perforin, interferon-γ (IFNγ), and tumor necrosis factor-α (TNFα), on antigen recognition when compared with WT1-TCR-engineered T cells still expressing their eTCR (eTCR+/+ WT1-TCR). CRISPR-Cas9 editing did not affect immunophenotypic characteristics or T cell activation and did not induce increased expression of inhibitory molecules. eTCR-/- WT1-TCR CD8+ CB-T cells showed effective migratory and killing capacity in cocultures with neoplastic cell lines and primary AML blasts, but did not show toxicity toward healthy cells., Conclusions: In summary, we show the feasibility of developing a potent CB-derived CD8+ T cell product targeting WT1, providing an option for post-transplant allogeneic immune cell therapy or as an off-the-shelf product, to prevent relapse and improve the clinical outcome of children with AML., Competing Interests: Competing interests: JK reports grants from Gadeta, Novartis, and Miltenyi Biotec and is the inventor of patents dealing with γδT cell-related aspects, as well as the cofounder and shareholder of Gadeta. ZS is an inventor of patents dealing with γδT cell-related aspects., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
16. Integrative single-cell expression and functional studies unravels a sensitization to cytarabine-based chemotherapy through HIF pathway inhibition in AML leukemia stem cells.
- Author
-
Velasco-Hernandez T, Trincado JL, Vinyoles M, Closa A, Martínez-Moreno A, Gutiérrez-Agüera F, Molina O, Rodríguez-Cortez VC, Ximeno-Parpal P, Fernández-Fuentes N, Petazzi P, Beneyto-Calabuig S, Velten L, Romecin P, Casquero R, Abollo-Jiménez F, de la Guardia RD, Lorden P, Bataller A, Lapillonne H, Stam RW, Vives S, Torrebadell M, Fuster JL, Bueno C, Sarry JE, Eyras E, Heyn H, and Menéndez P
- Abstract
Relapse remains a major challenge in the clinical management of acute myeloid leukemia (AML) and is driven by rare therapy-resistant leukemia stem cells (LSCs) that reside in specific bone marrow niches. Hypoxia signaling maintains cells in a quiescent and metabolically relaxed state, desensitizing them to chemotherapy. This suggests the hypothesis that hypoxia contributes to the chemoresistance of AML-LSCs and may represent a therapeutic target to sensitize AML-LSCs to chemotherapy. Here, we identify HIF
high and HIFlow specific AML subgroups (inv(16)/ t (8;21) and MLLr, respectively) and provide a comprehensive single-cell expression atlas of 119,000 AML cells and AML-LSCs in paired diagnostic-relapse samples from these molecular subgroups. The HIF/hypoxia pathway signature is attenuated in AML-LSCs compared with more differentiated AML cells but is more expressed than in healthy hematopoietic cells. Importantly, chemical inhibition of HIF cooperates with standard-of-care chemotherapy to impair AML growth and to substantially eliminate AML-LSCs in vitro and in vivo. These findings support the HIF pathway in the stem cell-driven drug resistance of AML and unravel avenues for combinatorial targeted and chemotherapy-based approaches to specifically eliminate AML-LSCs., Competing Interests: Pablo Menéndez is the founder of the spin‐off OneChain Immunotherapeutics, which has no connection with the present research. The other authors declare no conflict of interest., (© 2024 The Authors. HemaSphere published by John Wiley & Sons Ltd. on behalf of European Hematology Association.)- Published
- 2024
- Full Text
- View/download PDF
17. Modelling acquired resistance to DOT1L inhibition exhibits the adaptive potential of KMT2A-rearranged acute lymphoblastic leukemia.
- Author
-
Schneider P, Crump NT, Arentsen-Peters STCJM, Smith AL, Hagelaar R, Adriaanse FRS, Bos RS, de Jong A, Nierkens S, Koopmans B, Milne TA, Pieters R, and Stam RW
- Abstract
In KMT2A-rearranged acute lymphoblastic leukemia (ALL), an aggressive malignancy, oncogenic KMT2A-fusion proteins inappropriately recruit DOT1L to promote leukemogenesis, highlighting DOT1L as an attractive therapeutic target. Unfortunately, treatment with the first-in-class DOT1L inhibitor pinometostat eventually leads to non-responsiveness. To understand this we established acquired pinometostat resistance in pediatric KMT2A::AFF1
+ B-ALL cells. Interestingly, these cells became mostly independent of DOT1L-mediated H3K79 methylation, but still relied on the physical presence of DOT1L, HOXA9 and the KMT2A::AFF1 fusion. Moreover, these cells selectively lost the epigenetic regulation and expression of various KMT2A-fusion target genes such as PROM1/CD133, while other KMT2A::AFF1 target genes, including HOXA9 and CDK6 remained unaffected. Concomitantly, these pinometostat-resistant cells showed upregulation of several myeloid-associated genes, including CD33 and LILRB4/CD85k. Taken together, this model comprehensively shows the adaptive potential of KMT2A-rearranged ALL cells upon losing dependency on one of its main oncogenic properties., (© 2023. YUMED Inc. and BioMed Central Ltd.)- Published
- 2023
- Full Text
- View/download PDF
18. CRISPR-Cas9 Library Screening Identifies Novel Molecular Vulnerabilities in KMT2A -Rearranged Acute Lymphoblastic Leukemia.
- Author
-
Schneider P, Wander P, Arentsen-Peters STCJM, Vrenken KS, Rockx-Brouwer D, Adriaanse FRS, Hoeve V, Paassen I, Drost J, Pieters R, and Stam RW
- Subjects
- Humans, Gene Library, Transcription Factors, Cell Line, Antigens, Neoplasm, Neoplasm Proteins, CRISPR-Cas Systems, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
In acute lymphoblastic leukemia (ALL), chromosomal translocations involving the KMT2A gene represent highly unfavorable prognostic factors and most commonly occur in patients less than 1 year of age. Rearrangements of the KMT2A gene drive epigenetic changes that lead to aberrant gene expression profiles that strongly favor leukemia development. Apart from this genetic lesion, the mutational landscape of KMT2A -rearranged ALL is remarkably silent, providing limited insights for the development of targeted therapy. Consequently, identifying potential therapeutic targets often relies on differential gene expression, yet the inhibition of these genes has rarely translated into successful therapeutic strategies. Therefore, we performed CRISPR-Cas9 knock-out screens to search for genetic dependencies in KMT2A -rearranged ALL. We utilized small-guide RNA libraries directed against the entire human epigenome and kinome in various KMT2A -rearranged ALL, as well as wild-type KMT2A ALL cell line models. This screening approach led to the discovery of the epigenetic regulators ARID4B and MBD3 , as well as the receptor kinase BMPR2 as novel molecular vulnerabilities and attractive therapeutic targets in KMT2A -rearranged ALL.
- Published
- 2023
- Full Text
- View/download PDF
19. A convergent malignant phenotype in B-cell acute lymphoblastic leukemia involving the splicing factor SRRM1.
- Author
-
Closa A, Reixachs-Solé M, Fuentes-Fayos AC, Hayer KE, Melero JL, Adriaanse FRS, Bos RS, Torres-Diz M, Hunger SP, Roberts KG, Mullighan CG, Stam RW, Thomas-Tikhonenko A, Castaño JP, Luque RM, and Eyras E
- Abstract
A significant proportion of infant B-cell acute lymphoblastic leukemia (B-ALL) patients remains with a dismal prognosis due to yet undetermined mechanisms. We performed a comprehensive multicohort analysis of gene expression, gene fusions, and RNA splicing alterations to uncover molecular signatures potentially linked to the observed poor outcome. We identified 87 fusions with significant allele frequency across patients and shared functional impacts, suggesting common mechanisms across fusions. We further identified a gene expression signature that predicts high risk independently of the gene fusion background and includes the upregulation of the splicing factor SRRM1 . Experiments in B-ALL cell lines provided further evidence for the role of SRRM1 on cell survival, proliferation, and invasion. Supplementary analysis revealed that SRRM1 potentially modulates splicing events associated with poor outcomes through protein-protein interactions with other splicing factors. Our findings reveal a potential convergent mechanism of aberrant RNA processing that sustains a malignant phenotype independently of the underlying gene fusion and that could potentially complement current clinical strategies in infant B-ALL., (© The Author(s) 2022. Published by Oxford University Press on behalf of NAR Cancer.)
- Published
- 2022
- Full Text
- View/download PDF
20. A CRISPR/Cas9 engineered Mpl S504N mouse model recapitulates human myelofibrosis.
- Author
-
Adriaanse FRS, Kamens JL, Vogel P, Sakurada SM, Pruett-Miller SM, Stam RW, Michel Zwaan C, and Gruber TA
- Subjects
- Animals, CRISPR-Cas Systems, Disease Models, Animal, Humans, Mice, Receptors, Thrombopoietin genetics, Primary Myelofibrosis genetics
- Published
- 2022
- Full Text
- View/download PDF
21. Editorial: Harnessing chemotherapy resistance and development of novel therapeutic strategies for acute leukemia with KMT2A (MLL)-gene rearrangements.
- Author
-
Esposito MT, Hagström-Andersson A, Stam RW, and Bortoluzzi S
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
22. Hypoxic, glycolytic metabolism is a vulnerability of B-acute lymphoblastic leukemia-initiating cells.
- Author
-
Morris V, Wang D, Li Z, Marion W, Hughes T, Sousa P, Harada T, Sui SH, Naumenko S, Kalfon J, Sensharma P, Falchetti M, Vinicius da Silva R, Candelli T, Schneider P, Margaritis T, Holstege FCP, Pikman Y, Harris M, Stam RW, Orkin SH, Koehler AN, Shalek AK, North TE, Pimkin M, Daley GQ, Lummertz da Rocha E, and Rowe RG
- Subjects
- Glycolysis, Humans, Hypoxia, Leukemia, Myeloid, Acute metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
High-risk forms of B-acute lymphoblastic leukemia (B-ALL) remain a therapeutic challenge. Leukemia-initiating cells (LICs) self-renew and spark relapse and therefore have been the subject of intensive investigation; however, the properties of LICs in high-risk B-ALL are not well understood. Here, we use single-cell transcriptomics and quantitative xenotransplantation to understand LICs in MLL-rearranged (MLL-r) B-ALL. Compared with reported LIC frequencies in acute myeloid leukemia (AML), engraftable LICs in MLL-r B-ALL are abundant. Although we find that multipotent, self-renewing LICs are enriched among phenotypically undifferentiated B-ALL cells, LICs with the capacity to replenish the leukemic cellular diversity can emerge from more mature fractions. While inhibiting oxidative phosphorylation blunts blast proliferation, this intervention promotes LIC emergence. Conversely, inhibiting hypoxia and glycolysis impairs MLL-r B-ALL LICs, providing a therapeutic benefit in xenotransplantation systems. These findings provide insight into the aggressive nature of MLL-r B-ALL and provide a rationale for therapeutic targeting of hypoxia and glycolysis., Competing Interests: Declaration of interests G.Q.D. holds equity in companies pursuing anti-cancer treatments (including Epizyme and 28-7 Therapeutics) and patents related to cancer therapeutics. A.K.S. reports compensation for consulting and/or science advisory board (SAB) membership from Merck, Honeycomb Biotechnologies, Cellarity, Repertoire Immune Medicines, Ochre Bio, Third Rock Ventures, Hovione, Relation Therapeutics Limited, Empress Therapeutics, FL82, and Dahlia Biosciences., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
23. High-Throughput Drug Library Screening in Primary KMT2A -Rearranged Infant ALL Cells Favors the Identification of Drug Candidates That Activate P53 Signaling.
- Author
-
Wander P, Arentsen-Peters STCJM, Vrenken KS, Pinhanҫos SM, Koopmans B, Dolman MEM, Jones L, Garrido Castro P, Schneider P, Kerstjens M, Molenaar JJ, Pieters R, Zwaan CM, and Stam RW
- Abstract
KMT2A-rearranged acute lymphoblastic leukemia (ALL) in infants (<1 year of age) represents an aggressive type of childhood leukemia characterized by a poor clinical outcome with a survival chance of <50%. Implementing novel therapeutic approaches for these patients is a slow-paced and costly process. Here, we utilized a drug-repurposing strategy to identify potent drugs that could expeditiously be translated into clinical applications. We performed high-throughput screens of various drug libraries, comprising 4191 different (mostly FDA-approved) compounds in primary KMT2A-rearranged infant ALL patient samples (n = 2). The most effective drugs were then tested on non-leukemic whole bone marrow samples (n = 2) to select drugs with a favorable therapeutic index for bone marrow toxicity. The identified agents frequently belonged to several recurrent drug classes, including BCL-2, histone deacetylase, topoisomerase, microtubule, and MDM2/p53 inhibitors, as well as cardiac glycosides and corticosteroids. The in vitro efficacy of these drug classes was successfully validated in additional primary KMT2A-rearranged infant ALL samples (n = 7) and KMT2A-rearranged ALL cell line models (n = 5). Based on literature studies, most of the identified drugs remarkably appeared to lead to activation of p53 signaling. In line with this notion, subsequent experiments showed that forced expression of wild-type p53 in KMT2A-rearranged ALL cells rapidly led to apoptosis induction. We conclude that KMT2A-rearranged infant ALL cells are vulnerable to p53 activation, and that drug-induced p53 activation may represent an essential condition for successful treatment results. Moreover, the present study provides an attractive collection of approved drugs that are highly effective against KMT2A-rearranged infant ALL cells while showing far less toxicity towards non-leukemic bone marrow, urging further (pre)clinical testing.
- Published
- 2022
- Full Text
- View/download PDF
24. Identification and characterization of relapse-initiating cells in MLL-rearranged infant ALL by single-cell transcriptomics.
- Author
-
Candelli T, Schneider P, Garrido Castro P, Jones LA, Bodewes E, Rockx-Brouwer D, Pieters R, Holstege FCP, Margaritis T, and Stam RW
- Subjects
- Adult, Child, Child, Preschool, Female, Follow-Up Studies, Gene Expression Regulation, Leukemic, Humans, Infant, Infant, Newborn, Male, Neoplasm Recurrence, Local genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Prognosis, Survival Rate, Tumor Cells, Cultured, Biomarkers, Tumor genetics, Gene Rearrangement, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Neoplasm Recurrence, Local pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Single-Cell Analysis methods, Transcriptome
- Abstract
Infants with MLL-rearranged infant acute lymphoblastic leukemia (MLL-r iALL) undergo intense therapy to counter a highly aggressive malignancy with survival rates of only 30-40%. The majority of patients initially show therapy response, but in two-thirds of cases the leukemia returns, typically during treatment. The glucocorticoid drug prednisone is established as a major player in the treatment of leukemia and the in vivo response to prednisone monotreatment is currently the best indicator of risk for MLL-r iALL. We used two different single-cell RNA sequencing technologies to analyze the expression of a prednisone-dependent signature, derived from an independent study, in diagnostic bone marrow and peripheral blood biopsies. This allowed us to classify individual leukemic cells as either resistant or sensitive to treatment and show that quantification of these two groups can be used to better predict the occurrence of future relapse in individual patients. This work also sheds light on the nature of the therapy-resistant subpopulation of relapse-initiating cells. Leukemic cells associated with high relapse risk are characterized by basal activation of glucocorticoid response, smaller size, and a quiescent gene expression program with cell stemness properties. These results improve current risk stratification and elucidate leukemic therapy-resistant subpopulations at diagnosis., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
25. Favorable outcome of NUTM1-rearranged infant and pediatric B cell precursor acute lymphoblastic leukemia in a collaborative international study.
- Author
-
Boer JM, Valsecchi MG, Hormann FM, Antić Ž, Zaliova M, Schwab C, Cazzaniga G, Arfeuille C, Cavé H, Attarbaschi A, Strehl S, Escherich G, Imamura T, Ohki K, Grüber TA, Sutton R, Pastorczak A, Lammens T, Lambert F, Li CK, Carrillo de Santa Pau E, Hoffmann S, Möricke A, Harrison CJ, Den Boer ML, De Lorenzo P, Stam RW, Bergmann AK, and Pieters R
- Subjects
- Child, Female, Humans, Infant, Male, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma therapy, Prognosis, Survival Rate, Gene Rearrangement, Neoplasm Proteins genetics, Nuclear Proteins genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality
- Published
- 2021
- Full Text
- View/download PDF
26. Integrative methylome-transcriptome analysis unravels cancer cell vulnerabilities in infant MLL-rearranged B cell acute lymphoblastic leukemia.
- Author
-
Tejedor JR, Bueno C, Vinyoles M, Petazzi P, Agraz-Doblas A, Cobo I, Torres-Ruiz R, Bayón GF, Pérez RF, López-Tamargo S, Gutierrez-Agüera F, Santamarina-Ojeda P, Ramírez-Orellana M, Bardini M, Cazzaniga G, Ballerini P, Schneider P, Stam RW, Varela I, Fraga MF, Fernández AF, and Menéndez P
- Subjects
- Animals, Cell Proliferation drug effects, Cell Proliferation genetics, Core Binding Factor Alpha 2 Subunit genetics, CpG Islands, DNA Methylation, Epigenesis, Genetic, Epigenome, Gene Expression Profiling, Gene Expression Regulation, Leukemic, Humans, Infant, Mice, Mice, Inbred NOD, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Transcription Factor AP-1 antagonists & inhibitors, Transcription Factor AP-1 genetics, Transcription Factor AP-1 metabolism, Xenograft Model Antitumor Assays, Gene Rearrangement, B-Lymphocyte, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism
- Abstract
B cell acute lymphoblastic leukemia (B-ALL) is the most common childhood cancer. As predicted by its prenatal origin, infant B-ALL (iB-ALL) shows an exceptionally silent DNA mutational landscape, suggesting that alternative epigenetic mechanisms may substantially contribute to its leukemogenesis. Here, we have integrated genome-wide DNA methylome and transcriptome data from 69 patients with de novo MLL-rearranged leukemia (MLLr) and non-MLLr iB-ALL leukemia uniformly treated according to the Interfant-99/06 protocol. iB-ALL methylome signatures display a plethora of common and specific alterations associated with chromatin states related to enhancer and transcriptional control in normal hematopoietic cells. DNA methylation, gene expression, and gene coexpression network analyses segregated MLLr away from non-MLLr iB-ALL and identified a coordinated and enriched expression of the AP-1 complex members FOS and JUN and RUNX factors in MLLr iB-ALL, consistent with the significant enrichment of hypomethylated CpGs in these genes. Integrative methylome-transcriptome analysis identified consistent cancer cell vulnerabilities, revealed a robust iB-ALL-specific gene expression-correlating dmCpG signature, and confirmed an epigenetic control of AP-1 and RUNX members in reshaping the molecular network of MLLr iB-ALL. Finally, pharmacological inhibition or functional ablation of AP-1 dramatically impaired MLLr-leukemic growth in vitro and in vivo using MLLr-iB-ALL patient-derived xenografts, providing rationale for new therapeutic avenues in MLLr-iB-ALL.
- Published
- 2021
- Full Text
- View/download PDF
27. HDAC7 is a major contributor in the pathogenesis of infant t(4;11) proB acute lymphoblastic leukemia.
- Author
-
de Barrios O, Galaras A, Trincado JL, Azagra A, Collazo O, Meler A, Agraz-Doblas A, Bueno C, Ballerini P, Cazzaniga G, Stam RW, Varela I, De Lorenzo P, Valsecchi MG, Hatzis P, Menéndez P, and Parra M
- Subjects
- Humans, Infant, Myeloid-Lymphoid Leukemia Protein genetics, Translocation, Genetic genetics, Histone Deacetylases genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2021
- Full Text
- View/download PDF
28. Irinotecan Induces Disease Remission in Xenograft Mouse Models of Pediatric MLL -Rearranged Acute Lymphoblastic Leukemia.
- Author
-
Kerstjens M, Garrido Castro P, Pinhanços SS, Schneider P, Wander P, Pieters R, and Stam RW
- Abstract
Acute lymphoblastic leukemia (ALL) in infants (<1 year of age) remains one of the most aggressive types of childhood hematologic malignancy. The majority (~80%) of infant ALL cases are characterized by chromosomal translocations involving the MLL (or KMT2A ) gene, which confer highly dismal prognoses on current combination chemotherapeutic regimens. Hence, more adequate therapeutic strategies are urgently needed. To expedite clinical transition of potentially effective therapeutics, we here applied a drug repurposing approach by performing in vitro drug screens of (mostly) clinically approved drugs on a variety of human ALL cell line models. Out of 3685 compounds tested, the alkaloid drug Camptothecin (CPT) and its derivatives 10-Hydroxycamtothecin (10-HCPT) and 7-Ethyl-10-hydroxycamtothecin (SN-38: the active metabolite of the drug Irinotecan) appeared most effective at very low nanomolar concentrations in all ALL cell lines, including models of MLL -rearranged ALL ( n = 3). Although the observed in vitro anti-leukemic effects of Camptothecin and its derivatives certainly were not specific to MLL -rearranged ALL, we decided to further focus on this highly aggressive type of leukemia. Given that Irinotecan (the pro-drug of SN-38) has been increasingly used for the treatment of various pediatric solid tumors, we specifically chose this agent for further pre-clinical evaluation in pediatric MLL -rearranged ALL. Interestingly, shortly after engraftment, Irinotecan completely blocked leukemia expansion in mouse xenografts of a pediatric MLL -rearranged ALL cell line, as well as in two patient-derived xenograft (PDX) models of MLL -rearranged infant ALL. Also, from a more clinically relevant perspective, Irinotecan monotherapy was able to induce sustainable disease remissions in MLL -rearranged ALL xenotransplanted mice burdened with advanced leukemia. Taken together, our data demonstrate that Irinotecan exerts highly potent anti-leukemia effects against pediatric MLL -rearranged ALL, and likely against other, more favorable subtypes of childhood ALL as well.
- Published
- 2021
- Full Text
- View/download PDF
29. High-Throughput Screening Identifies Idasanutlin as a Resensitizing Drug for Venetoclax-Resistant Neuroblastoma Cells.
- Author
-
Vernooij L, Bate-Eya LT, Alles LK, Lee JY, Koopmans B, Jonus HC, Schubert NA, Schild L, Lelieveld D, Egan DA, Kerstjens M, Stam RW, Koster J, Goldsmith KC, Molenaar JJ, and Dolman MEM
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Humans, Mice, Proto-Oncogene Proteins c-mdm2 pharmacology, Pyrrolidines pharmacology, para-Aminobenzoates pharmacology, High-Throughput Screening Assays methods, Neuroblastoma drug therapy, Proto-Oncogene Proteins c-mdm2 therapeutic use, Pyrrolidines therapeutic use, para-Aminobenzoates therapeutic use
- Abstract
Neuroblastoma tumors frequently overexpress the anti-apoptotic protein B-cell lymphoma/leukemia 2 (BCL-2). We previously showed that treating BCL-2-dependent neuroblastoma cells with the BCL-2 inhibitor venetoclax results in apoptosis, but unfortunately partial therapy resistance is observed. The current study describes the identification of drugs capable of resensitizing venetoclax-resistant neuroblastoma cells to venetoclax. To examine these effects, venetoclax resistance was induced in BCL-2-dependent neuroblastoma cell lines KCNR and SJNB12 by continuous exposure to high venetoclax concentrations. Non-resistant and venetoclax-resistant neuroblastoma cell lines were exposed to a 209-compound library in the absence and presence of venetoclax to identify compounds that were more effective in the venetoclax-resistant cell lines under venetoclax pressure. Top hits were further validated in combination with venetoclax using BCL-2-dependent neuroblastoma model systems. Overall, high-throughput drug screening identified the MDM2 inhibitor idasanutlin as a promising resensitizing agent for venetoclax-resistant neuroblastoma cell lines. Idasanutlin treatment induced BAX-mediated apoptosis in venetoclax-resistant neuroblastoma cells in the presence of venetoclax, whereas it caused p21-mediated growth arrest in control cells. In vivo combination treatment showed tumor regression and superior efficacy over single-agent therapies in a BCL-2-dependent neuroblastoma cell line xenograft and a patient-derived xenograft. However, xenografts less dependent on BCL-2 were not sensitive to venetoclax-idasanutlin combination therapy. This study demonstrates that idasanutlin can overcome resistance to the BCL-2 inhibitor venetoclax in preclinical neuroblastoma model systems, which supports clinical development of a treatment strategy combining the two therapies., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
30. High-throughput drug screening reveals Pyrvinium pamoate as effective candidate against pediatric MLL-rearranged acute myeloid leukemia.
- Author
-
Wander P, Arentsen-Peters STCJM, Pinhanҫos SS, Koopmans B, Dolman MEM, Ariese R, Bos FL, Castro PG, Jones L, Schneider P, Navarro MG, Molenaar JJ, Rios AC, Zwaan CM, and Stam RW
- Abstract
Pediatric MLL-rearranged acute myeloid leukemia (AML) has a generally unfavorable outcome, primarily due to relapse and drug resistance. To overcome these difficulties, new therapeutic agents are urgently needed. Yet, implementing novel drugs for clinical use is a time-consuming, laborious, costly and high-risk process. Therefore, we applied a drug-repositioning strategy by screening drug libraries, comprised of >4000 compounds that are mostly FDA-approved, in a high-throughput format on primary MLL-rearranged AML cells. Here we identified pyrvinium pamoate (pyrvinium) as a novel candidate drug effective against MLL-rearranged AML, eliminating all cell viability at <1000 nM. Additional screening of identified drug hits on non-leukemic bone marrow samples, resulted in a decrease in cell viability of ∼50% at 1000 nM pyrvinium, suggesting a therapeutic window for targeting leukemic cells specifically. Validation of pyrvinium on an extensive panel of AML cell lines and primary AML samples showed comparable viabilities as the drug screen data, with pyrvinium achieving IC
50 values of <80 nM in these samples. Remarkably, pyrvinium also induced cell toxicity in primary MLL-AF10+ AML cells, an MLL-rearrangement associated with a poor outcome. While pyrvinium is able to inhibit the Wnt pathway in other diseases, this unlikely explains the efficacy we observed as β-catenin was not expressed in the AML cells tested. Rather, we show that pyrvinium co-localized with the mitochondrial stain in cells, and hence may act by inhibiting mitochondrial respiration. Overall, this study shows that pyrvinium is highly effective against MLL-rearranged AML in vitro, and therefore represents a novel potential candidate for further studies in MLL-rearranged AML., Competing Interests: Declaration of Competing Interest None., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
31. Preclinical Evaluation of Carfilzomib for Infant KMT2A -Rearranged Acute Lymphoblastic Leukemia.
- Author
-
Cheung LC, de Kraa R, Oommen J, Chua GA, Singh S, Hughes AM, Ferrari E, Ford J, Chiu SK, Stam RW, Kees UR, Malinge S, and Kotecha RS
- Abstract
Background: Infants with KMT2A -rearranged B-cell precursor acute lymphoblastic leukemia (ALL) have poor outcomes. There is an urgent need to identify novel agents to improve survival. Proteasome inhibition has emerged as a promising therapeutic strategy for several hematological malignancies. The aim of this study was to determine the preclinical efficacy of the selective proteasome inhibitor carfilzomib, for infants with KMT2A -rearranged ALL., Methods: Eight infant ALL cell lines were extensively characterized for immunophenotypic and cytogenetic features. In vitro cytotoxicity to carfilzomib was assessed using a modified Alamar Blue assay with cells in logarithmic growth. The Bliss Independence model was applied to determine synergy between carfilzomib and the nine conventional chemotherapeutic agents used to treat infants with ALL. Established xenograft models were used to identify the maximal tolerated dose of carfilzomib and determine in vivo efficacy., Results: Carfilzomib demonstrated low IC
50 concentrations within the nanomolar range (6.0-15.8 nm) across the panel of cell lines. Combination drug testing indicated in vitro synergy between carfilzomib and several conventional chemotherapeutic agents including vincristine, daunorubicin, dexamethasone, L-asparaginase, and 4-hydroperoxycyclophosphamide. In vivo assessment did not lead to a survival advantage for either carfilzomib monotherapy, when used to treat both low or high disease burden, or for carfilzomib in combination with multi-agent induction chemotherapy comprising of vincristine, dexamethasone, and L-asparaginase., Conclusions: Our study highlights that in vitro efficacy does not necessarily translate to benefit in vivo and emphasizes the importance of in vivo validation prior to suggesting an agent for clinical use. Whilst proteasome inhibitors have an important role to play in several hematological malignancies, our findings guard against prioritization of carfilzomib for treatment of KMT2A -rearranged infant ALL in the clinical setting., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Cheung, de Kraa, Oommen, Chua, Singh, Hughes, Ferrari, Ford, Chiu, Stam, Kees, Malinge and Kotecha.)- Published
- 2021
- Full Text
- View/download PDF
32. The clinical and biological characteristics of NUP98-KDM5A in pediatric acute myeloid leukemia.
- Author
-
Noort S, Wander P, Alonzo TA, Smith J, Ries RE, Gerbing RB, Dolman MEM, Locatelli F, Reinhardt D, Baruchel A, Stary J, Molenaar JJ, Stam RW, van den Heuvel-Eibrink MM, Zwaan MC, and Meshinchi S
- Subjects
- Child, Homeodomain Proteins genetics, Humans, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Retinoblastoma-Binding Protein 2, Translocation, Genetic, Leukemia, Myeloid, Acute genetics, Nuclear Pore Complex Proteins genetics
- Published
- 2021
- Full Text
- View/download PDF
33. Preclinical efficacy of gemcitabine in MLL-rearranged infant acute lymphoblastic leukemia.
- Author
-
Wander P, Cheung LC, Pinhanҫos SS, Jones L, Kerstjens M, Arentsen-Peters STCJM, Singh S, Chua GA, Castro PG, Schneider P, Dolman MEM, Koopmans B, Molenaar JJ, Pieters R, Zwaan CM, Kotecha RS, and Stam RW
- Subjects
- Animals, Antimetabolites, Antineoplastic therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Disease Management, Disease Models, Animal, Drug Evaluation, Preclinical, Genetic Predisposition to Disease, Humans, Infant, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Gemcitabine, Antimetabolites, Antineoplastic pharmacology, Deoxycytidine analogs & derivatives, Gene Rearrangement, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2020
- Full Text
- View/download PDF
34. Decitabine mildly attenuates MLL -rearranged acute lymphoblastic leukemia in vivo, and represents a poor chemo-sensitizer.
- Author
-
Schneider P, Castro PG, Pinhanços SM, Kerstjens M, van Roon EH, Essing AHW, Dolman MEM, Molenaar JJ, Pieters R, and Stam RW
- Abstract
MLL -rearranged acute lymphoblastic leukemia (ALL) represents a highly aggressive ALL subtype, characterized by aberrant DNA methylation patterns. DNA methyltransferase inhibitors, such as decitabine have previously been demonstrated to be effective in eradicating MLL -rearranged ALL cells in vitro. Here, we assessed the in vivo anti-leukemic potential of low-dose DNA methyltransferase inhibitor decitabine using a xenograft mouse model of human MLL -rearranged ALL. Furthermore, we explored whether prolonged exposure to low-dose decitabine could chemo-sensitize MLL -rearranged ALL cells toward conventional chemotherapy as well as other known epigenetic-based and anti-neoplastic compounds. Our data reveal that decitabine prolonged survival in xenograft mice of MLL -rearranged ALL by 8.5 days ( P = .0181), but eventually was insufficient to prevent leukemia out-growth, based on the examination of the MLLAF4 cell line SEM. Furthermore, we observe that prolonged pretreatment of low-dose decitabine mildly sensitized toward the conventional drugs prednisolone, vincristine, daunorubicin, asparaginase, and cytarabine in a panel of MLL -rearranged cell lines. Additionally, we assessed synergistic effects of decitabine with other epigenetic-based or anticancer drugs using high-throughput drug library screens. Validation of the top hits, including histone deacetylase inhibitor panobinostat, BCL2 inhibitor Venetoclax, MEK inhibitor pimasertib, and receptor tyrosine kinase foretinib, revealed additive and moderate synergistic effects for the combination of each drug together with decitabine in a cell line-dependent manner., Competing Interests: The authors declare that no conflict of interest exists., (© 2020 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
35. Partner independent fusion gene detection by multiplexed CRISPR-Cas9 enrichment and long read nanopore sequencing.
- Author
-
Stangl C, de Blank S, Renkens I, Westera L, Verbeek T, Valle-Inclan JE, González RC, Henssen AG, van Roosmalen MJ, Stam RW, Voest EE, Kloosterman WP, van Haaften G, and Monroe GR
- Subjects
- Cell Line, Tumor, High-Throughput Nucleotide Sequencing, Humans, Male, Neoplasms genetics, Sequence Analysis, DNA, CRISPR-Cas Systems genetics, Gene Fusion, Genetic Testing methods, Nanopore Sequencing, Neoplasms diagnosis
- Abstract
Fusion genes are hallmarks of various cancer types and important determinants for diagnosis, prognosis and treatment. Fusion gene partner choice and breakpoint-position promiscuity restricts diagnostic detection, even for known and recurrent configurations. Here, we develop FUDGE (FUsion Detection from Gene Enrichment) to accurately and impartially identify fusions. FUDGE couples target-selected and strand-specific CRISPR-Cas9 activity for fusion gene driver enrichment - without prior knowledge of fusion partner or breakpoint-location - to long read nanopore sequencing with the bioinformatics pipeline NanoFG. FUDGE has flexible target-loci choices and enables multiplexed enrichment for simultaneous analysis of several genes in multiple samples in one sequencing run. We observe on-average 665 fold breakpoint-site enrichment and identify nucleotide resolution fusion breakpoints within 2 days. The assay identifies cancer cell line and tumor sample fusions irrespective of partner gene or breakpoint-position. FUDGE is a rapid and versatile fusion detection assay for diagnostic pan-cancer fusion detection.
- Published
- 2020
- Full Text
- View/download PDF
36. NUTM1 is a recurrent fusion gene partner in B-cell precursor acute lymphoblastic leukemia associated with increased expression of genes on chromosome band 10p12.31-12.2.
- Author
-
Hormann FM, Hoogkamer AQ, Beverloo HB, Boeree A, Dingjan I, Wattel MM, Stam RW, Escherich G, Pieters R, den Boer ML, and Boer JM
- Subjects
- Child, Chromosome Banding, Chromosomes, Human, Female, Humans, Male, Gene Expression Regulation, Leukemic, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Nuclear Proteins biosynthesis, Nuclear Proteins genetics, Oncogene Proteins, Fusion biosynthesis, Oncogene Proteins, Fusion genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology
- Published
- 2019
- Full Text
- View/download PDF
37. FLT3 N676K drives acute myeloid leukemia in a xenograft model of KMT2A-MLLT3 leukemogenesis.
- Author
-
Hyrenius-Wittsten A, Pilheden M, Falqués-Costa A, Eriksson M, Sturesson H, Schneider P, Wander P, Garcia-Ruiz C, Liu J, Ågerstam H, Hultquist A, Lilljebjörn H, Stam RW, Järås M, and Hagström-Andersson AK
- Subjects
- Animals, Antigens, CD34 genetics, Gene Rearrangement genetics, Heterografts, Humans, Mice, Mice, Inbred NOD, Carcinogenesis genetics, Histone-Lysine N-Methyltransferase genetics, Leukemia, Myeloid, Acute genetics, Myeloid-Lymphoid Leukemia Protein genetics, Nuclear Proteins genetics, fms-Like Tyrosine Kinase 3 genetics
- Published
- 2019
- Full Text
- View/download PDF
38. Unraveling the cellular origin and clinical prognostic markers of infant B-cell acute lymphoblastic leukemia using genome-wide analysis.
- Author
-
Agraz-Doblas A, Bueno C, Bashford-Rogers R, Roy A, Schneider P, Bardini M, Ballerini P, Cazzaniga G, Moreno T, Revilla C, Gut M, Valsecchi MG, Roberts I, Pieters R, De Lorenzo P, Varela I, Menendez P, and Stam RW
- Subjects
- Biopsy, Bone Marrow metabolism, Chromosome Aberrations, Gene Expression Profiling, Gene Rearrangement, Genomic Instability, Histone-Lysine N-Methyltransferase genetics, Homeodomain Proteins genetics, Humans, In Situ Hybridization, Fluorescence, Mutation, Myeloid-Lymphoid Leukemia Protein genetics, Phosphatidylinositol 3-Kinases metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality, Prognosis, Signal Transduction, Survival Analysis, V(D)J Recombination, ras Proteins metabolism, Biomarkers, Tumor, Disease Susceptibility, Genome-Wide Association Study, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma etiology
- Abstract
B-cell acute lymphoblastic leukemia is the commonest childhood cancer. In infants, B-cell acute lymphoblastic leukemia remains fatal, especially in patients with t(4;11), present in ~80% of cases. The pathogenesis of t(4;11)/KMT2A-AFF1
+ (MLL-AF4+ ) infant B-cell acute lymphoblastic leukemia remains difficult to model, and the pathogenic contribution in cancer of the reciprocal fusions resulting from derivative translocated-chromosomes remains obscure. Here, "multi-layered" genome-wide analyses and validation were performed on a total of 124 de novo cases of infant B-cell acute lymphoblastic leukemia uniformly diagnosed and treated according to the Interfant 99/06 protocol. These patients showed the most silent mutational landscape reported so far for any sequenced pediatric cancer. Recurrent mutations were exclusively found in K-RAS and N-RAS , were subclonal and were frequently lost at relapse, despite a larger number of non-recurrent/non-silent mutations. Unlike non-MLL-rearranged B-cell acute lymphoblastic leukemias, B-cell receptor repertoire analysis revealed minor, non-expanded B-cell clones in t(4;11)+ infant B-cell acute lymphoblastic leukemia, and RNA-sequencing showed transcriptomic similarities between t(4;11)+ infant B-cell acute lymphoblastic leukemias and the most immature human fetal liver hematopoietic stem and progenitor cells, confirming a "pre-VDJ" fetal cellular origin for both t(4;11) and RASmut The reciprocal fusion AF4-MLL was expressed in only 45% (19/43) of the t(4;11)+ patients, and HOXA cluster genes are exclusively expressed in AF4-MLL -expressing patients. Importantly, AF4-MLL / HOXA -expressing patients had a significantly better 4-year event-free survival (62.4% vs 11.7%, P =0.001), and overall survival (73.7 vs 25.2%, P =0.016). AF4-MLL expression retained its prognostic significance when analyzed in a Cox model adjusting for risk stratification according to the Interfant-06 protocol based on age at diagnosis, white blood cell count and response to prednisone. This study has clinical implications for disease outcome and diagnostic risk-stratification of t(4;11)+ infant B-cell acute lymphoblastic leukemia., (Copyright© 2019 Ferrata Storti Foundation.)- Published
- 2019
- Full Text
- View/download PDF
39. A phase 1/2, open-label, dose-escalation study of midostaurin in children with relapsed or refractory acute leukaemia.
- Author
-
Zwaan CM, Söderhäll S, Brethon B, Luciani M, Rizzari C, Stam RW, Besse E, Dutreix C, Fagioli F, Ho PA, Dufour C, and Pieters R
- Subjects
- Adolescent, Child, Child, Preschool, Disease-Free Survival, Female, Follow-Up Studies, Humans, Infant, Leukemia, Myeloid, Acute metabolism, Male, Staurosporine administration & dosage, Staurosporine adverse effects, Survival Rate, Leukemia, Myeloid, Acute drug therapy, Staurosporine analogs & derivatives
- Published
- 2019
- Full Text
- View/download PDF
40. Ex vivo resistance in childhood acute lymphoblastic leukemia: Correlations between BCRP, MRP1, MRP4 and MRP5 ABC transporter expression and intracellular methotrexate polyglutamate accumulation.
- Author
-
Jaramillo AC, Cloos J, Lemos C, Stam RW, Kaspers GJL, Jansen G, and Peters GJ
- Subjects
- ATP-Binding Cassette Transporters genetics, Adolescent, Child, Child, Preschool, Cytoplasm drug effects, Cytoplasm metabolism, Drug Screening Assays, Antitumor, Female, Gene Expression Regulation, Leukemic drug effects, Humans, Infant, Infant, Newborn, Male, Methotrexate metabolism, Pilot Projects, Polyglutamic Acid metabolism, Primary Cell Culture, Tumor Cells, Cultured, ATP Binding Cassette Transporter, Subfamily G, Member 2 genetics, Drug Resistance, Neoplasm genetics, Methotrexate analogs & derivatives, Methotrexate therapeutic use, Multidrug Resistance-Associated Proteins genetics, Neoplasm Proteins genetics, Polyglutamic Acid analogs & derivatives, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
Chemoresistance is an important factor in the treatment failure of childhood acute lymphoblastic leukemia (ALL). One underlying mechanism of chemoresistance involves (over)expression of ATP-dependent drug efflux transporters such as multidrug resistance protein 1-5 (MRP1-5) and breast cancer resistance protein (BCRP), which can extrude the important antileukemia drug methotrexate (MTX). Survival of childhood ALL critically depends on the leukemic blasts' capacity for intracellular retention of MTX and MTX-polyglutamates. This pilot study assessed whether expression of MRP1, MRP4, MRP5 and BCRP (real-time PCR) in primary childhood ALL blasts (n = 23) correlated with ex vivo resistance to MTX (assayed by in situ thymidylate synthase inhibition assay (TSIA)), ex vivo accumulation of (radioactive) MTX polyglutamates, and patient survival. Results show that high MRP4 expression is correlated with ex vivo MTX resistance assayed by TSIA (P = 0.01). Moreover, elevated MRP4 and BCRP expression correlated with lower accumulation of MTX-PGs (P = 0.004 and P = 0.03, respectively). Combined high expression of BCRP and MRP4 even further impacted reduced MTX-PG accumulation (P = 0.02). Overall survival was lower (P logrank = 0.04) in children with ALL cells which featured a relatively high expression of both BCRP and MRP4 transporters. These results underscore the impact of high drug efflux transporter expression, notably MRP4 and BCRP, in diminished MTX response in childhood ALL., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
41. Correction: NG2 antigen is involved in leukemia invasiveness and central nervous system infiltration in MLL-rearranged infant B-ALL.
- Author
-
Prieto C, López-Millán B, Roca-Ho H, Stam RW, Romero-Moya D, Rodríguez-Baena FJ, Sanjuan-Pla A, Ayllón V, Ramírez M, Bardini M, De Lorenzo P, Valsecchi MG, Stanulla M, Iglesias M, Ballerini P, Carcaboso ÁM, Mora J, Locatelli F, Bertaina A, Padilla L, Rodríguez-Manzaneque JC, Bueno C, and Menéndez P
- Abstract
The original version of this Article contained an error in the spelling of the author Juan Carlos Rodriguez-Manzaneque, which was incorrectly given as J Carlos Rodríguez-Manzaneque. This has now been corrected in both the PDF and HTML versions of the Article.
- Published
- 2018
- Full Text
- View/download PDF
42. Antileukemic Efficacy of BET Inhibitor in a Preclinical Mouse Model of MLL-AF4 + Infant ALL.
- Author
-
Bardini M, Trentin L, Rizzo F, Vieri M, Savino AM, Garrido Castro P, Fazio G, Van Roon EHJ, Kerstjens M, Smithers N, Prinjha RK, Te Kronnie G, Basso G, Stam RW, Pieters R, Biondi A, and Cazzaniga G
- Subjects
- Animals, Humans, Mice, Mice, Inbred NOD, Mice, Mutant Strains, Mice, SCID, Transcriptome, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
MLL -rearranged acute lymphoblastic leukemia (ALL) occurring in infants is a rare but very aggressive leukemia, typically associated with a dismal prognosis. Despite the development of specific therapeutic protocols, infant patients with MLL -rearranged ALL still suffer from a low cure rate. At present, novel therapeutic approaches are urgently needed. Recently, the use of small molecule inhibitors targeting the epigenetic regulators of the MLL complex emerged as a promising strategy for the development of a targeted therapy. Herein, we have investigated the effects of bromodomain and extra-terminal (BET) function abrogation in a preclinical mouse model of MLL-AF4
+ infant ALL using the BET inhibitor I-BET151. We reported that I-BET151 is able to arrest the growth of MLL-AF4+ leukemic cells in vitro , by blocking cell division and rapidly inducing apoptosis. Treatment with I-BET151 in vivo impairs the leukemic engraftment of patient-derived primary samples and lower the disease burden in mice. I-BET151 affects the transcriptional profile of MLL -rearranged ALL through the deregulation of BRD4, HOXA7/HOXA9 , and RUNX1 gene networks. Moreover, I-BET151 treatment sensitizes glucocorticoid-resistant MLL -rearranged cells to prednisolone in vitro and is more efficient when used in combination with HDAC inhibitors, both in vitro and in vivo Given the aggressiveness of the disease, the failure of the current therapies and the lack of an ultimate cure, this study paves the way for the use of BET inhibitors to treat MLL -rearranged infant ALL for future clinical applications. Mol Cancer Ther; 17(8); 1705-16. ©2018 AACR ., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
43. Trametinib inhibits RAS -mutant MLL -rearranged acute lymphoblastic leukemia at specific niche sites and reduces ERK phosphorylation in vivo .
- Author
-
Kerstjens M, Pinhancos SS, Castro PG, Schneider P, Wander P, Pieters R, and Stam RW
- Subjects
- Animals, Cell Line, Tumor, Gene Rearrangement, Heterografts, Humans, MAP Kinase Signaling System, Mice, Phosphorylation drug effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnostic imaging, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Tumor Burden drug effects, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Pyridones pharmacology, Pyrimidinones pharmacology, ras Proteins genetics
- Published
- 2018
- Full Text
- View/download PDF
44. NG2 antigen is involved in leukemia invasiveness and central nervous system infiltration in MLL-rearranged infant B-ALL.
- Author
-
Prieto C, López-Millán B, Roca-Ho H, Stam RW, Romero-Moya D, Rodríguez-Baena FJ, Sanjuan-Pla A, Ayllón V, Ramírez M, Bardini M, De Lorenzo P, Valsecchi MG, Stanulla M, Iglesias M, Ballerini P, Carcaboso ÁM, Mora J, Locatelli F, Bertaina A, Padilla L, Rodríguez-Manzaneque JC, Bueno C, and Menéndez P
- Abstract
Mixed-lineage leukemia (MLL)-rearranged (MLLr) infant B-cell acute lymphoblastic leukemia (iMLLr-B-ALL) has a dismal prognosis and is associated with a pro-B/mixed phenotype, therapy refractoriness and frequent central nervous system (CNS) disease/relapse. Neuron-glial antigen 2 (NG2) is specifically expressed in MLLr leukemias and is used in leukemia immunophenotyping because of its predictive value for MLLr acute leukemias. NG2 is involved in melanoma metastasis and brain development; however, its role in MLL-mediated leukemogenesis remains elusive. Here we evaluated whether NG2 distinguishes leukemia-initiating/propagating cells (L-ICs) and/or CNS-infiltrating cells (CNS-ICs) in iMLLr-B-ALL. Clinical data from the Interfant cohort of iMLLr-B-ALL demonstrated that high NG2 expression associates with lower event-free survival, higher number of circulating blasts and more frequent CNS disease/relapse. Serial xenotransplantation of primary MLL-AF4
+ leukemias indicated that NG2 is a malleable marker that does not enrich for L-IC or CNS-IC in iMLLr-B-All. However, NG2 expression was highly upregulated in blasts infiltrating extramedullar hematopoietic sites and CNS, and specific blockage of NG2 resulted in almost complete loss of engraftment. Indeed, gene expression profiling of primary blasts and primografts revealed a migratory signature of NG2+ blasts. This study provides new insights on the biology of NG2 in iMLLr-B-ALL and suggests NG2 as a potential therapeutic target to reduce the risk of CNS disease/relapse and to provide safer CNS-directed therapies for iMLLr-B-ALL.- Published
- 2018
- Full Text
- View/download PDF
45. The HDAC inhibitor panobinostat (LBH589) exerts in vivo anti-leukaemic activity against MLL-rearranged acute lymphoblastic leukaemia and involves the RNF20/RNF40/WAC-H2B ubiquitination axis.
- Author
-
Garrido Castro P, van Roon EHJ, Pinhanços SS, Trentin L, Schneider P, Kerstjens M, Te Kronnie G, Heidenreich O, Pieters R, and Stam RW
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Cell Death drug effects, Cell Death genetics, Epigenesis, Genetic drug effects, Epigenesis, Genetic genetics, Gene Rearrangement genetics, Heterografts drug effects, Heterografts metabolism, Histone Deacetylases metabolism, Histones genetics, Humans, Mice, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Ubiquitin-Protein Ligases genetics, Ubiquitination genetics, Gene Rearrangement drug effects, Histone Deacetylase Inhibitors pharmacology, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Panobinostat pharmacology, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Ubiquitination drug effects
- Abstract
MLL-rearranged acute lymphoblastic leukaemia (ALL) represents an aggressive malignancy in infants (<1 year of age), associated with poor outcome. Current treatment intensification is not further possible, and novel therapy strategies are needed. Notably, MLL-rearranged ALL is characterised by a strongly deregulated epigenome and shows sensitivity to epigenetic perturbators. Here we demonstrate the in vivo efficacy of the histone deacetylase inhibitor panobinostat (LBH589) using xenograft mouse models of MLL-rearranged ALL. Panobinostat monotherapy showed strong anti-leukaemic effects, extending survival and reducing overall disease burden. Comprehensive molecular analyses in vitro showed that this anti-leukaemic activity involves depletion of H2B ubiquitination via suppression of the RNF20/RNF40/WAC E3 ligase complex; a pivotal pathway for MLL-rearranged leukaemic maintenance. Knockdown of WAC phenocopied loss of H2B ubiquitination and concomitant cell death induction. These combined data demonstrate that panobinostat cross-inhibits multiple epigenetic pathways, ultimately contributing to its highly efficacious targeting of MLL-rearranged ALL.
- Published
- 2018
- Full Text
- View/download PDF
46. MEK inhibition is a promising therapeutic strategy for MLL-rearranged infant acute lymphoblastic leukemia patients carrying RAS mutations.
- Author
-
Kerstjens M, Driessen EM, Willekes M, Pinhanços SS, Schneider P, Pieters R, and Stam RW
- Subjects
- Apoptosis drug effects, Cell Proliferation drug effects, Drug Resistance, Neoplasm genetics, Humans, Infant, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Translocation, Genetic, Tumor Cells, Cultured, Gene Rearrangement, Histone-Lysine N-Methyltransferase genetics, MAP Kinase Kinase 1 antagonists & inhibitors, Mutation genetics, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Protein Kinase Inhibitors therapeutic use, ras Proteins genetics
- Abstract
Acute lymphoblastic leukemia (ALL) in infants is an aggressive malignancy with a poor clinical outcome, and is characterized by translocations of the Mixed Lineage Leukemia (MLL) gene. Previously, we identified RAS mutations in 14-24% of infant ALL patients, and showed that the presence of a RAS mutation decreased the survival chances even further. We hypothesized that targeting the RAS signaling pathway could be a therapeutic strategy for RAS-mutant infant ALL patients. Here we show that the MEK inhibitors Trametinib, Selumetinib and MEK162 severely impair primary RAS-mutant MLL-rearranged infant ALL cells in vitro. While all RAS-mutant samples were sensitive to MEK inhibitors, we found both sensitive and resistant samples among RAS-wildtype cases. We confirmed enhanced RAS pathway signaling in RAS-mutant samples, but found no apparent downstream over-activation in the wildtype samples. However, we did confirm that MEK inhibitors reduced p-ERK levels, and induced apoptosis in the RAS-mutant MLL-rearranged ALL cells. Finally, we show that MEK inhibition synergistically enhances prednisolone sensitivity, both in RAS-mutant and RAS-wildtype cells. In conclusion, MEK inhibition represents a promising therapeutic strategy for MLL-rearranged ALL patients harboring RAS mutations, while patients without RAS mutations may benefit through prednisolone sensitization.
- Published
- 2017
- Full Text
- View/download PDF
47. Updates in the biology and therapy for infant acute lymphoblastic leukemia.
- Author
-
Guest EM and Stam RW
- Subjects
- Combined Modality Therapy, Epigenesis, Genetic, Genetic Testing, Humans, Infant, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Prognosis, Antineoplastic Agents therapeutic use, Biomarkers, Tumor genetics, Hematopoietic Stem Cell Transplantation, Molecular Targeted Therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Purpose of Review: The prognosis for infants less than 12 months of age who are diagnosed with acute lymphoblastic leukemia (ALL) remains poor despite overall advances in the treatment of childhood ALL. In this review, we highlight the recent advances in the understanding of the pathogenesis of infant ALL and discuss opportunities for translating these findings into clinical trials., Recent Findings: Infant ALL can be divided into two major disease types, defined by the presence or absence of KMT2A (MLL) rearrangement (KMT2A-R). Recent molecular profiling studies have found that infant ALL with KMT2A-R is an epigenomic disease that lacks other somatic driver mutations. Strategies to intensify therapy have not improved survival for infants with KMT2A-R ALL. In contrast, infant ALL without KMT2A-R is more similar to ALL of older children and survival has improved modestly with intensification of chemotherapy. Discovery of clonal molecular markers that predict chemoresistance will allow further risk classification and development of novel treatment strategies. Modern clinical trials are integrating molecularly targeted therapies into the treatment of infant ALL., Summary: Advances in molecular profiling and integration of targeted therapy have the potential to reduce toxicity and improve survival for infants with ALL.
- Published
- 2017
- Full Text
- View/download PDF
48. Network-based expression analysis reveals key genes related to glucocorticoid resistance in infant acute lymphoblastic leukemia.
- Author
-
Mousavian Z, Nowzari-Dalini A, Stam RW, Rahmatallah Y, and Masoudi-Nejad A
- Subjects
- Gene Regulatory Networks, Histone-Lysine N-Methyltransferase genetics, Humans, Infant, Myeloid-Lymphoid Leukemia Protein genetics, Antineoplastic Agents therapeutic use, Drug Resistance, Neoplasm genetics, Gene Expression Profiling methods, Glucocorticoids therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Purpose: Despite vast improvements that have been made in the treatment of children with acute lymphoblastic leukemia (ALL), the majority of infant ALL patients (~80 %, < 1 year of age) that carry a chromosomal translocation involving the mixed lineage leukemia (MLL) gene shows a poor response to chemotherapeutic drugs, especially glucocorticoids (GCs), which are essential components of all current treatment regimens. Although addressed in several studies, the mechanism(s) underlying this phenomenon have remained largely unknown. A major drawback of most previous studies is their primary focus on individual genes, thereby neglecting the putative significance of inter-gene correlations. Here, we aimed at studying GC resistance in MLL-rearranged infant ALL patients by inferring an associated module of genes using co-expression network analysis. The implications of newly identified candidate genes with associations to other well-known relevant genes from the same module, or with associations to known transcription factor or microRNA interactions, were substantiated using literature data., Methods: A weighted gene co-expression network was constructed to identify gene modules associated with GC resistance in MLL-rearranged infant ALL patients. Significant gene ontology (GO) terms and signaling pathways enriched in relevant modules were used to provide guidance towards which module(s) consisted of promising candidates suitable for further analysis., Results: Through gene co-expression network analysis a novel set of genes (module) related to GC-resistance was identified. The presence in this module of the S100 and ANXA genes, both well-known biomarkers for GC resistance in MLL-rearranged infant ALL, supports its validity. Subsequent gene set net correlation analyses of the novel module provided further support for its validity by showing that the S100 and ANXA genes act as 'hub' genes with potentially major regulatory roles in GC sensitivity, but having lost this role in the GC resistant phenotype. The detected module implicates new genes as being candidates for further analysis through associations with known GC resistance-related genes., Conclusions: From our data we conclude that available systems biology approaches can be employed to detect new candidate genes that may provide further insights into drug resistance of MLL-rearranged infant ALL cases. Such approaches complement conventional gene-wise approaches by taking putative functional interactions between genes into account.
- Published
- 2017
- Full Text
- View/download PDF
49. Development Refractoriness of MLL-Rearranged Human B Cell Acute Leukemias to Reprogramming into Pluripotency.
- Author
-
Muñoz-López A, Romero-Moya D, Prieto C, Ramos-Mejía V, Agraz-Doblas A, Varela I, Buschbeck M, Palau A, Carvajal-Vergara X, Giorgetti A, Ford A, Lako M, Granada I, Ruiz-Xivillé N, Rodríguez-Perales S, Torres-Ruíz R, Stam RW, Fuster JL, Fraga MF, Nakanishi M, Cazzaniga G, Bardini M, Cobo I, Bayon GF, Fernandez AF, Bueno C, and Menendez P
- Subjects
- Animals, Biomarkers, Cell Line, Transformed, Cell Line, Tumor, Cluster Analysis, DNA Methylation, Gene Expression, Gene Expression Profiling, Hematopoietic Stem Cells metabolism, Heterografts, Humans, Mice, Myeloid Progenitor Cells metabolism, Oncogene Proteins, Fusion genetics, Phenotype, Precursor Cells, B-Lymphoid metabolism, Transcriptome, Translocation, Genetic, Cell Transdifferentiation genetics, Cellular Reprogramming, Gene Rearrangement, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Myeloid-Lymphoid Leukemia Protein genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Induced pluripotent stem cells (iPSCs) are a powerful tool for disease modeling. They are routinely generated from healthy donors and patients from multiple cell types at different developmental stages. However, reprogramming leukemias is an extremely inefficient process. Few studies generated iPSCs from primary chronic myeloid leukemias, but iPSC generation from acute myeloid or lymphoid leukemias (ALL) has not been achieved. We attempted to generate iPSCs from different subtypes of B-ALL to address the developmental impact of leukemic fusion genes. OKSM(L)-expressing mono/polycistronic-, retroviral/lentiviral/episomal-, and Sendai virus vector-based reprogramming strategies failed to render iPSCs in vitro and in vivo. Addition of transcriptomic-epigenetic reprogramming "boosters" also failed to generate iPSCs from B cell blasts and B-ALL lines, and when iPSCs emerged they lacked leukemic fusion genes, demonstrating non-leukemic myeloid origin. Conversely, MLL-AF4-overexpressing hematopoietic stem cells/B progenitors were successfully reprogrammed, indicating that B cell origin and leukemic fusion gene were not reprogramming barriers. Global transcriptome/DNA methylome profiling suggested a developmental/differentiation refractoriness of MLL-rearranged B-ALL to reprogramming into pluripotency., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
50. Immunophenotypic analysis and quantification of B-1 and B-2 B cells during human fetal hematopoietic development.
- Author
-
Bueno C, van Roon EH, Muñoz-López A, Sanjuan-Pla A, Juan M, Navarro A, Stam RW, and Menendez P
- Subjects
- Fetus, Humans, Immunophenotyping, B-Lymphocyte Subsets cytology, Hematopoiesis immunology, Hematopoietic System embryology, Immune System embryology
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.