360 results on '"Kuiper, R.P."'
Search Results
2. A Common Genomic Denominator for Neuroblastoma and Differentiated Thyroid Carcinoma? A Case Series in Children
- Author
-
Clement, S.C., primary, Koster, J., additional, Kuiper, R.P., additional, Tytgat, G.A.M., additional, Ebus, M.E., additional, Schild, L., additional, Gilissen, C., additional, Zwijnenburg, D.A., additional, Versteeg, R., additional, Molenaar, J.J., additional, and van Santen, H.M., additional
- Published
- 2022
- Full Text
- View/download PDF
3. Using reverse genetics to pave the way to patient tailored therapies for pediatric Acute lymphoblastic leukemia - Finding the (metabolic) Achilles’ heel of high-risk ALL subtypes
- Author
-
Geurts van Kessel, A.H.M., Kuiper, R.P., Leeuwen, F.N. van, Meer, L.T. van der, Butler, M.B., Geurts van Kessel, A.H.M., Kuiper, R.P., Leeuwen, F.N. van, Meer, L.T. van der, and Butler, M.B.
- Abstract
Radboud University, 01 juli 2022, Promotor : Geurts van Kessel, A.H.M. Co-promotores : Kuiper, R.P., Leeuwen, F.N. van, Meer, L.T. van der, Item does not contain fulltext
- Published
- 2022
4. Clonal dynamics in pediatric B-cell precursor acute lymphoblastic leukemia with very early relapse
- Author
-
Antić, Ž., Yu, J., Bornhauser, B.C., Lelieveld, S.H., Ham, C.G. van der, Reijmersdal, S.V. van, Morgado, L., Elitzur, S., Bourquin, J.P., Cazzaniga, G., Eckert, C., Camós, M., Sutton, R., Cavé, H., Moorman, A.V., Sonneveld, E., Geurts van Kessel, A.H.M., Leeuwen, F.N. van, Hoogerbrugge, P.M., Waanders, E., Kuiper, R.P, Antić, Ž., Yu, J., Bornhauser, B.C., Lelieveld, S.H., Ham, C.G. van der, Reijmersdal, S.V. van, Morgado, L., Elitzur, S., Bourquin, J.P., Cazzaniga, G., Eckert, C., Camós, M., Sutton, R., Cavé, H., Moorman, A.V., Sonneveld, E., Geurts van Kessel, A.H.M., Leeuwen, F.N. van, Hoogerbrugge, P.M., Waanders, E., and Kuiper, R.P
- Abstract
Item does not contain fulltext, 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.
- Published
- 2022
5. In Response to: Pediatric Myelodysplastic Syndrome with Germline RRAS Mutation: Expanding the Phenotype of RASopathies
- Author
-
Engelen, N, Diets, I.J., Bresters, D., Bergen, J.C. van den, Vrancken, A., Kuiper, R.P, Jongmans, M.C.J., Engelen, N, Diets, I.J., Bresters, D., Bergen, J.C. van den, Vrancken, A., Kuiper, R.P, and Jongmans, M.C.J.
- Abstract
Item does not contain fulltext
- Published
- 2022
6. Return of the clones: Mechanisms driving clonal evolution and relapse development in pediatric Acute Lymphoblastic Leukemia
- Author
-
Holstege, F.C.P., Kuiper, R.P., Antić, Željko, Holstege, F.C.P., Kuiper, R.P., and Antić, Željko
- Published
- 2022
7. Renal tumors in children and the role of (epi)genetic predisposition
- Author
-
van den Heuvel-Eibrink, M.M., Jongmans, M.C.J., Kuiper, R.P., Hol, Janna Aldegonda, van den Heuvel-Eibrink, M.M., Jongmans, M.C.J., Kuiper, R.P., and Hol, Janna Aldegonda
- Published
- 2022
8. A Common Genomic Denominator for Neuroblastoma and Differentiated Thyroid Carcinoma? A Case Series in Children
- Author
-
Clement, S.C., Koster, J., Kuiper, R.P., Tytgat, G.A., Ebus, M.E., Schild, L., Gilissen, C., Zwijnenburg, D.A., Versteeg, R., Molenaar, J.J., Santen, H.M. van, Clement, S.C., Koster, J., Kuiper, R.P., Tytgat, G.A., Ebus, M.E., Schild, L., Gilissen, C., Zwijnenburg, D.A., Versteeg, R., Molenaar, J.J., and Santen, H.M. van
- Abstract
Item does not contain fulltext
- Published
- 2022
9. Germline MBD4 deficiency causes a multi-tumor predisposition syndrome
- Author
-
Palles, C., West, H.D., Chew, E., Galavotti, S., Flensburg, C., Grolleman, J.E., Jansen, E.A.M., Curley, H., Chegwidden, L., Arbe-Barnes, E.H., Lander, N., Truscott, R., Pagan, J., Bajel, A., Sherwood, K., Martin, L., Thomas, H, Georgiou, D., Fostira, F., Goldberg, Y., Adams, D.J., Biezen, S.A.M. van der, Christie, M., Clendenning, M., Thomas, L.E., Deltas, C., Dimovski, A.J., Dymerska, D., Lubinski, J., Mahmood, K., Post, R.S. van der, Sanders, M., Weitz, J., Taylor, J.C., Turnbull, C., Vreede, L., Wezel, T. van, Whalley, C., Arnedo-Pac, C., Caravagna, G., Cross, W., Chubb, D., Frangou, A., Gruber, A.J., Kinnersley, B., Noyvert, B., Church, D., Graham, T., Houlston, R., Lopez-Bigas, N., Sottoriva, A., Wedge, D., Jenkins, Mark A., Kuiper, R.P., Roberts, A.W., Cheadle, J.P., Ligtenberg, M.J.L., Hoogerbrugge, N., Koelzer, V.H., Rivas, A.D., Winship, I.M., Ponte, C.R., Buchanan, D.D., Power, D.G., Green, A., Tomlinson, I.P., Sampson, J.R., Majewski, I.J., Voer, R.M. de, Palles, C., West, H.D., Chew, E., Galavotti, S., Flensburg, C., Grolleman, J.E., Jansen, E.A.M., Curley, H., Chegwidden, L., Arbe-Barnes, E.H., Lander, N., Truscott, R., Pagan, J., Bajel, A., Sherwood, K., Martin, L., Thomas, H, Georgiou, D., Fostira, F., Goldberg, Y., Adams, D.J., Biezen, S.A.M. van der, Christie, M., Clendenning, M., Thomas, L.E., Deltas, C., Dimovski, A.J., Dymerska, D., Lubinski, J., Mahmood, K., Post, R.S. van der, Sanders, M., Weitz, J., Taylor, J.C., Turnbull, C., Vreede, L., Wezel, T. van, Whalley, C., Arnedo-Pac, C., Caravagna, G., Cross, W., Chubb, D., Frangou, A., Gruber, A.J., Kinnersley, B., Noyvert, B., Church, D., Graham, T., Houlston, R., Lopez-Bigas, N., Sottoriva, A., Wedge, D., Jenkins, Mark A., Kuiper, R.P., Roberts, A.W., Cheadle, J.P., Ligtenberg, M.J.L., Hoogerbrugge, N., Koelzer, V.H., Rivas, A.D., Winship, I.M., Ponte, C.R., Buchanan, D.D., Power, D.G., Green, A., Tomlinson, I.P., Sampson, J.R., Majewski, I.J., and Voer, R.M. de
- Abstract
Contains fulltext : 251996.pdf (Publisher’s version ) (Open Access), We report an autosomal recessive, multi-organ tumor predisposition syndrome, caused by bi-allelic loss-of-function germline variants in the base excision repair (BER) gene MBD4. We identified five individuals with bi-allelic MBD4 variants within four families and these individuals had a personal and/or family history of adenomatous colorectal polyposis, acute myeloid leukemia, and uveal melanoma. MBD4 encodes a glycosylase involved in repair of G:T mismatches resulting from deamination of 5'-methylcytosine. The colorectal adenomas from MBD4-deficient individuals showed a mutator phenotype attributable to mutational signature SBS1, consistent with the function of MBD4. MBD4-deficient polyps harbored somatic mutations in similar driver genes to sporadic colorectal tumors, although AMER1 mutations were more common and KRAS mutations less frequent. Our findings expand the role of BER deficiencies in tumor predisposition. Inclusion of MBD4 in genetic testing for polyposis and multi-tumor phenotypes is warranted to improve disease management.
- Published
- 2022
10. TRIM28 variants and Wilms' tumour predisposition
- Author
-
Hol, J.A., Diets, I.J., Krijger, R.R. de, Heuvel-Eibrink, M.M. van den, Jongmans, M.C.J., Kuiper, R.P., Hol, J.A., Diets, I.J., Krijger, R.R. de, Heuvel-Eibrink, M.M. van den, Jongmans, M.C.J., and Kuiper, R.P.
- Abstract
Item does not contain fulltext, TRIM28 was recently identified as a Wilms' tumour (WT) predisposition gene, with germline pathogenic variants identified in around 1% of isolated and 8% of familial WT cases. TRIM28 variants are associated with epithelial WT, but the presence of other tumour components or anaplasia does not exclude the presence of a germline or somatic TRIM28 variant. In children with WT, TRIM28 acts as a classical tumour suppressor gene, with both alleles generally disrupted in the tumour. Therefore, loss of TRIM28 (KAP1/TIF1beta) protein expression in tumour tissue by immunohistochemistry is an effective strategy to identify patients carrying pathogenic TRIM28 variants. TRIM28 is a ubiquitously expressed corepressor that binds transcription factors in a context-, species-, and cell-type-specific manner to control the expression of genes and transposable elements during embryogenesis and cellular differentiation. In this review, we describe the inheritance patterns, histopathological and clinical features of TRIM28-associated WT, as well as potential underlying mechanisms of tumourigenesis during embryonic kidney development. Recognizing germline TRIM28 variants in patients with WT can enable counselling, genetic testing, and potential early detection of WT in other children in the family. A further exploration of TRIM28-associated WT will help to unravel the diverse and complex mechanisms underlying WT development. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2021
11. Multiclonal complexity of pediatric acute lymphoblastic leukemia and the prognostic relevance of subclonal mutations
- Author
-
Antic, Zeljko, Yu, J., Reijmersdal, S.V. van, Dijk, A.H.A. van, Dekker, Linde, Segerink, Wouter H., Leeuwen, Frank N. van, Geurts van Kessel, A.H.M., Waanders, Esme, Kuiper, R.P., Antic, Zeljko, Yu, J., Reijmersdal, S.V. van, Dijk, A.H.A. van, Dekker, Linde, Segerink, Wouter H., Leeuwen, Frank N. van, Geurts van Kessel, A.H.M., Waanders, Esme, and Kuiper, R.P.
- Abstract
Contains fulltext : 242555.pdf (Publisher’s version ) (Open Access)
- Published
- 2021
12. BTK inhibition sensitizes acute lymphoblastic leukemia to asparaginase by suppressing the amino acid response pathway
- Author
-
Butler, M, Ingen Schenau, D.S. van, Yu, J., Jenni, S., Dobay, M.P., Hagelaar, R., Vervoort, B.M.T., Tee, T.M., Hoff, F.W., Meijerink, J.P., Kornblau, S.M., Bornhauser, B., Bourquin, J.P., Kuiper, R.P., Meer, L.T. van der, Leeuwen, F.N. van, Butler, M, Ingen Schenau, D.S. van, Yu, J., Jenni, S., Dobay, M.P., Hagelaar, R., Vervoort, B.M.T., Tee, T.M., Hoff, F.W., Meijerink, J.P., Kornblau, S.M., Bornhauser, B., Bourquin, J.P., Kuiper, R.P., Meer, L.T. van der, and Leeuwen, F.N. van
- Abstract
Item does not contain fulltext, Asparaginase (ASNase) therapy has been a mainstay of acute lymphoblastic leukemia (ALL) protocols for decades and shows promise in the treatment of a variety of other cancers. To improve the efficacy of ASNase treatment, we used a CRISPR/Cas9-based screen to identify actionable signaling intermediates that improve the response to ASNase. Both genetic inactivation of Bruton's tyrosine kinase (BTK) and pharmacological inhibition by the BTK inhibitor ibrutinib strongly synergize with ASNase by inhibiting the amino acid response pathway, a mechanism involving c-Myc-mediated suppression of GCN2 activity. This synthetic lethal interaction was observed in 90% of patient-derived xenografts, regardless of the genomic subtype. Moreover, ibrutinib substantially improved ASNase treatment response in a murine PDX model. Hence, ibrutinib may be used to enhance the clinical efficacy of ASNase in ALL. This trial was registered at www.clinicaltrials.gov as # NCT02884453.
- Published
- 2021
13. Genetic childhood cancer predisposition. Towards better detection and understanding
- Author
-
Hoogerbrugge-van der Linden, N., Jongmans, M.C.J., Kuiper, R.P., Diets, I.J., Hoogerbrugge-van der Linden, N., Jongmans, M.C.J., Kuiper, R.P., and Diets, I.J.
- Abstract
Radboud University, 10 september 2021, Promotor : Hoogerbrugge-van der Linden, N. Co-promotores : Jongmans, M.C.J., Kuiper, R.P., Contains fulltext : 216546.pdf (Publisher’s version ) (Open Access)
- Published
- 2021
14. Optical genome mapping identifies a germline retrotransposon insertion in SMARCB1 in two siblings with atypical teratoid rhabdoid tumors
- Author
-
Sabatella, M., Mantere, Tuomo, Waanders, E., Neveling, K., Mensenkamp, A.R., Dijk, F. van, Hehir, J.Y., Derks, R, Kwint, M.P., O'Gorman, L., Martins, M., Gidding, C.E.M., Lequin, M.H., Kusters, B., Wesseling, P., Nelen, M.R., Biegel, J.A., Hoischen, A., Jongmans, M.C.J., Kuiper, R.P., Sabatella, M., Mantere, Tuomo, Waanders, E., Neveling, K., Mensenkamp, A.R., Dijk, F. van, Hehir, J.Y., Derks, R, Kwint, M.P., O'Gorman, L., Martins, M., Gidding, C.E.M., Lequin, M.H., Kusters, B., Wesseling, P., Nelen, M.R., Biegel, J.A., Hoischen, A., Jongmans, M.C.J., and Kuiper, R.P.
- Abstract
Item does not contain fulltext, In a subset of pediatric cancers, a germline cancer predisposition is highly suspected based on clinical and pathological findings, but genetic evidence is lacking, which hampers genetic counseling and predictive testing in the families involved. We describe a family with two siblings born from healthy parents who were both neonatally diagnosed with atypical teratoid rhabdoid tumor (ATRT). This rare and aggressive pediatric tumor is associated with biallelic inactivation of SMARCB1, and in 30% of the cases, a predisposing germline mutation is involved. Whereas the tumors of both siblings showed loss of expression of SMARCB1 and acquired homozygosity of the locus, whole exome and whole genome sequencing failed to identify germline or somatic SMARCB1 pathogenic mutations. We therefore hypothesized that the insertion of a pathogenic repeat-rich structure might hamper its detection, and we performed optical genome mapping (OGM) as an alternative strategy to identify structural variation in this locus. Using this approach, an insertion of ~2.8 kb within intron 2 of SMARCB1 was detected. Long-range PCR covering this region remained unsuccessful, but PacBio HiFi genome sequencing identified this insertion to be a SINE-VNTR-Alu, subfamily E (SVA-E) retrotransposon element, which was present in a mosaic state in the mother. This SVA-E insertion disrupts correct splicing of the gene, resulting in loss of a functional allele. This case demonstrates the power of OGM and long-read sequencing to identify genomic variations in high-risk cancer-predisposing genes that are refractory to detection with standard techniques, thereby completing the clinical and molecular diagnosis of such complex cases and greatly improving counseling and surveillance of the families involved. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2021
15. Identifying and characterizing hereditary polyposis and colorectal cancer: Mutational signatures of defective base excision repair
- Author
-
Hoogerbrugge-van der Linden, N., Ligtenberg, M.J.L., Voer, R.M. de, Kuiper, R.P., Grolleman, J.E., Hoogerbrugge-van der Linden, N., Ligtenberg, M.J.L., Voer, R.M. de, Kuiper, R.P., and Grolleman, J.E.
- Abstract
Radboud University, 09 november 2021, Promotores : Hoogerbrugge-van der Linden, N., Ligtenberg, M.J.L. Co-promotores : Voer, R.M. de, Kuiper, R.P., Contains fulltext : 239215.pdf (Publisher’s version ) (Open Access)
- Published
- 2021
16. Challenges of Neoantigen Targeting in Lynch Syndrome and Constitutional Mismatch Repair Deficiency Syndrome
- Author
-
Abidi, A., Gorris, M.A.J., Brennan, E., Jongmans, M.C.J., Weijers, D.D., Kuiper, R.P., Voer, R.M. de, Hoogerbrugge, N., Schreibelt, G., Vries, I.J.M. de, Abidi, A., Gorris, M.A.J., Brennan, E., Jongmans, M.C.J., Weijers, D.D., Kuiper, R.P., Voer, R.M. de, Hoogerbrugge, N., Schreibelt, G., and Vries, I.J.M. de
- Abstract
Contains fulltext : 234983.pdf (Publisher’s version ) (Open Access), Lynch syndrome (LS) and constitutional mismatch repair deficiency (CMMRD) are hereditary disorders characterised by a highly increased risk of cancer development. This is due to germline aberrations in the mismatch repair (MMR) genes, which results in a high mutational load in tumours of these patients, including insertions and deletions in genes bearing microsatellites. This generates microsatellite instability and cause reading frameshifts in coding regions that could lead to the generation of neoantigens and opens up avenues for neoantigen targeting immune therapies prophylactically and therapeutically. However, major obstacles need to be overcome, such as the heterogeneity in tumour formation within and between LS and CMMRD patients, which results in considerable variability in the genes targeted by mutations, hence challenging the choice of suitable neoantigens. The machine-learning methods such as NetMHC and MHCflurry that predict neoantigen- human leukocyte antigen (HLA) binding affinity provide little information on other aspects of neoantigen presentation. Immune escape mechanisms that allow MMR-deficient cells to evade surveillance combined with the resistance to immune checkpoint therapy make the neoantigen targeting regimen challenging. Studies to delineate shared neoantigen profiles across patient cohorts, precise HLA binding algorithms, additional therapies to counter immune evasion and evaluation of biomarkers that predict the response of these patients to immune checkpoint therapy are warranted.
- Published
- 2021
17. Selection criteria for assembling a pediatric cancer predisposition syndrome gene panel
- Author
-
Byrjalsen, A., Diets, I.J., Bakhuizen, J., Hansen, T.V., Schmiegelow, K., Gerdes, A.M., Stoltze, U., Kuiper, R.P, Merks, J.H., Wadt, K., Jongmans, M., Byrjalsen, A., Diets, I.J., Bakhuizen, J., Hansen, T.V., Schmiegelow, K., Gerdes, A.M., Stoltze, U., Kuiper, R.P, Merks, J.H., Wadt, K., and Jongmans, M.
- Abstract
Contains fulltext : 244086.pdf (Publisher’s version ) (Open Access), Increasing use of genomic sequencing enables standardized screening of all childhood cancer predisposition syndromes (CPS) in children with cancer. Gene panels currently used often include adult-onset CPS genes and genes without substantial evidence linking them to cancer predisposition. We have developed criteria to select genes relevant for childhood-onset CPS and assembled a gene panel for use in children with cancer. We applied our criteria to 381 candidate genes, which were selected through two in-house panels (n = 338), a literature search (n = 39), and by assessing two Genomics England's PanelApp panels (n = 4). We developed evaluation criteria that determined a gene's eligibility for inclusion on a childhood-onset CPS gene panel. These criteria assessed (1) relevance in childhood cancer by a minimum of five childhood cancer patients reported carrying a pathogenic variant in the gene and (2) evidence supporting a causal relation between variants in this gene and cancer development. 138 genes fulfilled the criteria. In this study we have developed criteria to compile a childhood cancer predisposition gene panel which might ultimately be used in a clinical setting, regardless of the specific type of childhood cancer. This panel will be evaluated in a prospective study. The panel is available on (pediatric-cancer-predisposition-genepanel.nl) and will be regularly updated.
- Published
- 2021
18. Natural gene therapy in hematopoietic disorders: GATA too
- Author
-
Jongmans, M.C.J., Kuiper, R.P., Jongmans, M.C.J., and Kuiper, R.P.
- Abstract
Contains fulltext : 225479.pdf (Publisher’s version ) (Open Access)
- Published
- 2020
19. Renal cell carcinoma in young FH mutation carriers: case series and review of the literature
- Author
-
Hol, J.A., Jongmans, M.C.J., Littooij, A.S., Krijger, R.R. de, Kuiper, R.P, Harssel, J.J. van, Mensenkamp, A.R., Simons, M., Tytgat, G.A., Heuvel-Eibrink, M.M. van den, Grotel, M. van, Hol, J.A., Jongmans, M.C.J., Littooij, A.S., Krijger, R.R. de, Kuiper, R.P, Harssel, J.J. van, Mensenkamp, A.R., Simons, M., Tytgat, G.A., Heuvel-Eibrink, M.M. van den, and Grotel, M. van
- Abstract
Contains fulltext : 218591.pdf (Publisher’s version ) (Open Access), Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC) is an autosomal dominant syndrome caused by heterozygous pathogenic germline variants in the fumarate hydratase (FH) gene. It is characterized by cutaneous and uterine leiomyomas and an increased risk of developing renal cell carcinoma (RCC), which is usually adult-onset. HLRCC-related RCC tends to be aggressive and can metastasize even when the primary tumor is small. Data on children and adolescents are scarce. Herein, we report two patients from unrelated Dutch families, with HLRCC-related RCC at the ages of 15 and 18 years, and a third patient with an FH mutation and complex renal cysts at the age of 13. Both RCC's were localized and successfully resected, and careful MRI surveillance was initiated to monitor the renal cysts. One of the patients with RCC subsequently developed an ovarian Leydig cell tumor. A review of the literature identified 10 previously reported cases of HLRCC-related RCC in patients aged younger than 20 years, five of them presenting with metastatic disease. These data emphasize the importance of recognizing HLRCC in young patients to enable early detection of RCC, albeit rare. They support the recommendations from the 2014 consensus guideline, in which genetic testing for FH mutations, and renal MRI surveillance, is advised for HLRCC family members from the age of 8-10 years onwards.
- Published
- 2020
20. Clonal Relationship Between Lichen Sclerosus, Differentiated Vulvar Intra-epithelial Neoplasia and Non HPV-related Vulvar Squamous Cell Carcinoma
- Author
-
Pouwer, A.W., Einden, L.C.G. van den, Linden, M. van der, Hehir-Kwa, J.Y., Yu, Jiangyan, Hendriks, K.M., Kamping, E.J., Massuger, L.F.A.G., Bulten, J., Tilborg, A.G. van, Hullu, J.A. de, Kuiper, R.P., Pouwer, A.W., Einden, L.C.G. van den, Linden, M. van der, Hehir-Kwa, J.Y., Yu, Jiangyan, Hendriks, K.M., Kamping, E.J., Massuger, L.F.A.G., Bulten, J., Tilborg, A.G. van, Hullu, J.A. de, and Kuiper, R.P.
- Abstract
Contains fulltext : 217314.pdf (Publisher’s version ) (Open Access)
- Published
- 2020
21. Ribosomal protein gene RPL9 variants can differentially impair ribosome function and cellular metabolism
- Author
-
Lezzerini, M., Penzo, M., O'Donohue, M.F., Vieira, C. Marques Dos San, Saby, M., Elfrink, H.L., Diets, I.J., Hesse, A.M., Coute, Y., Gastou, M., Nin-Velez, A., Nikkels, P.G., Olson, A.N., Zonneveld-Huijssoon, E., Jongmans, M.C.J., Zhang, G., Weeghel, M. van, Houtkooper, R.H., Wlodarski, M.W., Kuiper, R.P, Bierings, M.B., Bosch, J, Leblanc, T., Montanaro, L., Dinman, J.D., Costa, L., Gleizes, P.E., Macinnes, A.W., Lezzerini, M., Penzo, M., O'Donohue, M.F., Vieira, C. Marques Dos San, Saby, M., Elfrink, H.L., Diets, I.J., Hesse, A.M., Coute, Y., Gastou, M., Nin-Velez, A., Nikkels, P.G., Olson, A.N., Zonneveld-Huijssoon, E., Jongmans, M.C.J., Zhang, G., Weeghel, M. van, Houtkooper, R.H., Wlodarski, M.W., Kuiper, R.P, Bierings, M.B., Bosch, J, Leblanc, T., Montanaro, L., Dinman, J.D., Costa, L., Gleizes, P.E., and Macinnes, A.W.
- Abstract
Contains fulltext : 218593.pdf (publisher's version ) (Open Access), Variants in ribosomal protein (RP) genes drive Diamond-Blackfan anemia (DBA), a bone marrow failure syndrome that can also predispose individuals to cancer. Inherited and sporadic RP gene variants are also linked to a variety of phenotypes, including malignancy, in individuals with no anemia. Here we report an individual diagnosed with DBA carrying a variant in the 5'UTR of RPL9 (uL6). Additionally, we report two individuals from a family with multiple cancer incidences carrying a RPL9 missense variant. Analysis of cells from these individuals reveals that despite the variants both driving pre-rRNA processing defects and 80S monosome reduction, the downstream effects are remarkably different. Cells carrying the 5'UTR variant stabilize TP53 and impair the growth and differentiation of erythroid cells. In contrast, ribosomes incorporating the missense variant erroneously read through UAG and UGA stop codons of mRNAs. Metabolic profiles of cells carrying the 5'UTR variant reveal an increased metabolism of amino acids and a switch from glycolysis to gluconeogenesis while those of cells carrying the missense variant reveal a depletion of nucleotide pools. These findings indicate that variants in the same RP gene can drive similar ribosome biogenesis defects yet still have markedly different downstream consequences and clinical impacts.
- Published
- 2020
22. Glucocorticoid Resistant Pediatric Acute Lymphoblastic Leukemia Samples Display Altered Splicing Profile and Vulnerability to Spliceosome Modulation
- Author
-
Sciarrillo, R., Wojtuszkiewicz, A., Kooi, I.E., Leon, L.G., Sonneveld, E., Kuiper, R.P., Jansen, G, Giovannetti, E., Kaspers, G.J., Cloos, J., Sciarrillo, R., Wojtuszkiewicz, A., Kooi, I.E., Leon, L.G., Sonneveld, E., Kuiper, R.P., Jansen, G, Giovannetti, E., Kaspers, G.J., and Cloos, J.
- Abstract
Contains fulltext : 220784.pdf (publisher's version ) (Open Access), Glucocorticoid (GC) resistance is a crucial determinant of inferior response to chemotherapy in pediatric acute lymphoblastic leukemia (ALL); however, molecular mechanisms underlying this phenomenon are poorly understood. Deregulated splicing is a common feature of many cancers, which impacts drug response and constitutes an attractive therapeutic target. Therefore, the aim of the current study was to characterize global splicing profiles associated with GC resistance and determine whether splicing modulation could serve as a novel therapeutic option for GC-resistant patients. To this end, 38 primary ALL samples were profiled using RNA-seq-based differential splicing analysis. The impact of splicing modulators was investigated in GC-resistant leukemia cell lines and primary leukemic specimens. Our findings revealed, for the first time, markedly distinct splicing landscapes in ALL samples of B-cell precursor (BCP)-ALL and T-ALL lineages. Differential splicing events associated with GC resistance were involved in RNA processing, a direct response to GCs, survival signaling, apoptosis, cell cycle regulation and energy metabolism. Furthermore, our analyses showed that GC-resistant ALL cell lines and primary samples are sensitive to splicing modulation, alone and in combination with GC. Together, these findings suggest that aberrant splicing is associated with GC resistance and splicing modulators deserve further interest as a novel treatment option for GC-resistant patients.
- Published
- 2020
23. The Game of clones in pediatric acute lymphoblastic leukemia. Mechanisms underlying relapse development
- Author
-
Geurts van Kessel, A.H.M., Kuiper, R.P., Waanders, E., Yu, J., Geurts van Kessel, A.H.M., Kuiper, R.P., Waanders, E., and Yu, J.
- Abstract
Radboud University, 03 november 2020, Promotor : Geurts van Kessel, A.H.M. Co-promotores : Kuiper, R.P., Waanders, E., Contains fulltext : 225167 .pdf (publisher's version ) (Open Access)
- Published
- 2020
24. Monoallelic NTHL1 Loss-of-Function Variants and Risk of Polyposis and Colorectal Cancer
- Author
-
Elsayed, F.A., Grolleman, J.E., Ragunathan, Abiramy, Buchanan, Daniel D., Hoogerbrugge, N., Kuiper, R.P., Ligtenberg, M.J.L., Wezel, Tom van, Voer, R.M. de, Elsayed, F.A., Grolleman, J.E., Ragunathan, Abiramy, Buchanan, Daniel D., Hoogerbrugge, N., Kuiper, R.P., Ligtenberg, M.J.L., Wezel, Tom van, and Voer, R.M. de
- Abstract
Contains fulltext : 228713.pdf (Publisher’s version ) (Open Access)
- Published
- 2020
25. Glucocorticoid resistant pediatric acute lymphoblastic leukemia samples display altered splicing profile and vulnerability to spliceosome modulation
- Author
-
Sciarrillo, R. (Rocco), Wojtuszkiewicz, A. (Anna), Kooi, I.E. (Irsan E.), Leon, L.G. (Leticia), Sonneveld, E. (Edwin), Kuiper, R.P. (Roland), Jansen, G. (Gert), Giovannetti, E. (Elisa), Kaspers, G.J.L. (Gertjan), Cloos, J. (Jacqueline), Sciarrillo, R. (Rocco), Wojtuszkiewicz, A. (Anna), Kooi, I.E. (Irsan E.), Leon, L.G. (Leticia), Sonneveld, E. (Edwin), Kuiper, R.P. (Roland), Jansen, G. (Gert), Giovannetti, E. (Elisa), Kaspers, G.J.L. (Gertjan), and Cloos, J. (Jacqueline)
- Abstract
Glucocorticoid (GC) resistance is a crucial determinant of inferior response to chemotherapy in pediatric acute lymphoblastic leukemia (ALL); however, molecular mechanisms underlying this phenomenon are poorly understood. Deregulated splicing is a common feature of many cancers, which impacts drug response and constitutes an attractive therapeutic target. Therefore, the aim of the current study was to characterize global splicing profiles associated with GC resistance and determine whether splicing modulation could serve as a novel therapeutic option for GC-resistant patients. To this end, 38 primary ALL samples were profiled using RNA-seq-based differential splicing analysis. The impact of splicing modulators was investigated in GC-resistant leukemia cell lines and primary leukemic specimens. Our findings revealed, for the first time, markedly distinct splicing landscapes in ALL samples of B-cell precursor (BCP)-ALL and T-ALL lineages. Differential splicing events associated with GC resistance were involved in RNA processing, a direct response to GCs, survival signaling, apoptosis, cell cycle regulation and energy metabolism. Furthermore, our analyses showed that GC-resistant ALL cell lines and primary samples are sensitive to splicing modulation, alone and in combination with GC. Together, these findings suggest that aberrant splicing is associated with GC resistance and splicing modulators deserve further interest as a novel treatment option for GC-resistant patients.
- Published
- 2020
- Full Text
- View/download PDF
26. Mutational landscape and patterns of clonal evolution in relapsed pediatric acute lymphoblastic leukemia
- Author
-
Waanders, E., Gu, Z., Dobson, S.M., Antić, Ž., Crawford, J.C., Ma, X., Edmonson, M.N., Payne-Turner, D., Vorst, M. van de, Jongmans, M.C.J., McGuire, I., Zhou, X., Wang, J, Shi, L., Pounds, S., Pei, D., Cheng, C., Song, G., Fan, Y., Shao, Y., Rusch, M., McCastlain, K., Yu, J., Boxtel, R. van, Blokzijl, F., Iacobucci, I., Roberts, K.G., Wen, J., Wu, G., Ma, J, Easton, J., Neale, G., Olsen, S.R., Nichols, K.E., Pui, C.H., Zhang, J., Evans, W.E., Relling, M.V., Yang, J.J., Thomas, P.G., Dick, J.E., Kuiper, R.P., Mullighan, C.G., Waanders, E., Gu, Z., Dobson, S.M., Antić, Ž., Crawford, J.C., Ma, X., Edmonson, M.N., Payne-Turner, D., Vorst, M. van de, Jongmans, M.C.J., McGuire, I., Zhou, X., Wang, J, Shi, L., Pounds, S., Pei, D., Cheng, C., Song, G., Fan, Y., Shao, Y., Rusch, M., McCastlain, K., Yu, J., Boxtel, R. van, Blokzijl, F., Iacobucci, I., Roberts, K.G., Wen, J., Wu, G., Ma, J, Easton, J., Neale, G., Olsen, S.R., Nichols, K.E., Pui, C.H., Zhang, J., Evans, W.E., Relling, M.V., Yang, J.J., Thomas, P.G., Dick, J.E., Kuiper, R.P., and Mullighan, C.G.
- Abstract
Item does not contain fulltext, Relapse of acute lymphoblastic leukemia (ALL) remains a leading cause of childhood death. Prior studies have shown clonal mutations at relapse often arise from relapse-fated subclones that exist at diagnosis. However, the genomic landscape, evolutionary trajectories and mutational mechanisms driving relapse are incompletely understood. In an analysis of 92 cases of relapsed childhood ALL, incorporating multimodal DNA and RNA sequencing, deep digital mutational tracking and xenografting to formally define clonal structure, we identify 50 significant targets of mutation with distinct patterns of mutational acquisition or enrichment. CREBBP, NOTCH1, and Ras signaling mutations rose from diagnosis subclones, whereas variants in NCOR2, USH2A and NT5C2 were exclusively observed at relapse. Evolutionary modeling and xenografting demonstrated that relapse-fated clones were minor (50%), major (27%) or multiclonal (18%) at diagnosis. Putative second leukemias, including those with lineage shift, were shown to most commonly represent relapse from an ancestral clone rather than a truly independent second primary leukemia. A subset of leukemias prone to repeated relapse exhibited hypermutation driven by at least three distinct mutational processes, resulting in heightened neoepitope burden and potential vulnerability to immunotherapy. Finally, relapse-driving sequence mutations were detected prior to relapse using deep digital PCR at levels comparable to orthogonal approaches to monitor levels of measurable residual disease. These results provide a genomic framework to anticipate and circumvent relapse by earlier detection and targeting of relapse-fated clones.
- Published
- 2020
27. Upfront Treatment Influences the Composition of Genetic Alterations in Relapsed Pediatric B-Cell Precursor Acute Lymphoblastic Leukemia
- Author
-
Yu, J., Waanders, E., Reijmersdal, S.V. van, Antic, Z., Bosbeek, C.M. van, Sonneveld, E., Groot, H. de, Fiocco, M., Geurts van Kessel, A.H.M., Leeuwen, F.N. van, Pieters, R., Hoogerbrugge, P.M., Kuiper, R.P, Yu, J., Waanders, E., Reijmersdal, S.V. van, Antic, Z., Bosbeek, C.M. van, Sonneveld, E., Groot, H. de, Fiocco, M., Geurts van Kessel, A.H.M., Leeuwen, F.N. van, Pieters, R., Hoogerbrugge, P.M., and Kuiper, R.P
- Abstract
Contains fulltext : 218617.pdf (publisher's version ) (Open Access), Genomic alterations in relapsed B-cell precursor acute lymphoblastic leukemia (BCP-ALL) may provide insight into the role of specific genomic events in relapse development. Along this line, comparisons between the spectrum of alterations in relapses that arise in different upfront treatment protocols may provide valuable information on the association between the tumor genome, protocol components and outcome. Here, we performed a comprehensive characterization of relapsed BCP-ALL cases that developed in the context of 3 completed Dutch upfront studies, ALL8, ALL9, and ALL10. In total, 123 pediatric BCP-ALL relapses and 77 paired samples from primary diagnosis were analyzed for alterations in 22 recurrently affected genes. We found pronounced differences in relapse alterations between the 3 studies. Specifically, CREBBP mutations were observed predominantly in relapses after treatment with ALL8 and ALL10 which, in the latter group, were all detected in medium risk-treated patients. IKZF1 alterations were enriched 2.2-fold (p = 0.01) and 2.9-fold (p < 0.001) in ALL8 and ALL9 relapses compared to diagnosis, respectively, whereas no significant enrichment was found for relapses that were observed after treatment with ALL10. Furthermore, IKZF1 deletions were more frequently preserved from a major clone at diagnosis in relapses after ALL9 compared to relapses after ALL8 and ALL10 (p = 0.03). These data are in line with previous studies showing that the prognostic value of IKZF1 deletions differs between upfront protocols and is particularly strong in the ALL9 regimen. In conclusion, our data reveal a correlation between upfront treatment and the genetic composition of relapsed BCP-ALL.
- Published
- 2020
28. NTHL1 and MUTYH polyposis syndromes: two sides of the same coin?
- Author
-
Weren, R.D.A., Ligtenberg, M.J.L., Geurts van Kessel, A., Voer, R.M. de, Hoogerbrugge, N., and Kuiper, R.P.
- Subjects
Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] - Abstract
Item does not contain fulltext It is now well established that germline genomic aberrations can underlie high-penetrant familial polyposis and colorectal cancer syndromes, but a genetic cause has not yet been found for the major proportion of patients with polyposis. Since next-generation sequencing has become widely accessible, several novel, but rare, high-penetrant risk factors for adenomatous polyposis have been identified, all operating in pathways responsible for genomic maintenance and DNA repair. One of these is the base excision repair pathway. In addition to the well-established role of the DNA glycosylase gene MUTYH, biallelic mutations in which predispose to MUTYH-associated polyposis, a second DNA glycosylase gene, NTHL1, has recently been associated with adenomatous polyposis and a high colorectal cancer risk. Both recessive polyposis syndromes are associated with increased risks for several other cancer types as well, but the spectrum of benign and malignant tumours in individuals with biallelic NTHL1 mutations was shown to be broader; hence the name NTHL1-associated tumour syndrome. Colorectal tumours encountered in patients with these syndromes show unique, clearly distinct mutational signatures that may facilitate the identification of these syndromes. On the basis of the prevalence of pathogenic MUTYH and NTHL1 variants in the normal population, we estimate that the frequency of the novel NTHL1-associated tumour syndrome is five times lower than that of MUTYH-associated polyposis. Copyright (c) 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2018
29. De Novo and Inherited Pathogenic Variants in KDM3B Cause Intellectual Disability, Short Stature, and Facial Dysmorphism
- Author
-
Diets, I.J., Donk, R. van der, Baltrunaite, K., Waanders, E., Reijnders, M.R.F., Dingemans, A.J., Pfundt, R.P., Vulto-van Silfhout, A.T., Wiel, L.J.M. van de, Gilissen, C.F., Thevenon, J., Perrin, L., Afenjar, A., Nava, C., Keren, B., Bartz, S., Peri, B., Beunders, G., Verbeek, N., Gassen, K. van, Thiffault, I., Cadieux-Dion, M., Huerta-Saenz, L., Wagner, M., Konstantopoulou, V., Vodopiutz, J., Griese, M., Boel, A., Callewaert, B., Brunner, H.G., Kleefstra, T., Hoogerbrugge, N., Vries, B.B. de, Hwa, V., Dauber, A., Hehir-Kwa, J.Y., Kuiper, R.P, Jongmans, M.C.J., Diets, I.J., Donk, R. van der, Baltrunaite, K., Waanders, E., Reijnders, M.R.F., Dingemans, A.J., Pfundt, R.P., Vulto-van Silfhout, A.T., Wiel, L.J.M. van de, Gilissen, C.F., Thevenon, J., Perrin, L., Afenjar, A., Nava, C., Keren, B., Bartz, S., Peri, B., Beunders, G., Verbeek, N., Gassen, K. van, Thiffault, I., Cadieux-Dion, M., Huerta-Saenz, L., Wagner, M., Konstantopoulou, V., Vodopiutz, J., Griese, M., Boel, A., Callewaert, B., Brunner, H.G., Kleefstra, T., Hoogerbrugge, N., Vries, B.B. de, Hwa, V., Dauber, A., Hehir-Kwa, J.Y., Kuiper, R.P, and Jongmans, M.C.J.
- Abstract
Contains fulltext : 202646.pdf (publisher's version ) (Open Access), By using exome sequencing and a gene matching approach, we identified de novo and inherited pathogenic variants in KDM3B in 14 unrelated individuals and three affected parents with varying degrees of intellectual disability (ID) or developmental delay (DD) and short stature. The individuals share additional phenotypic features that include feeding difficulties in infancy, joint hypermobility, and characteristic facial features such as a wide mouth, a pointed chin, long ears, and a low columella. Notably, two individuals developed cancer, acute myeloid leukemia and Hodgkin lymphoma, in childhood. KDM3B encodes for a histone demethylase and is involved in H3K9 demethylation, a crucial part of chromatin modification required for transcriptional regulation. We identified missense and truncating variants, suggesting that KDM3B haploinsufficiency is the underlying mechanism for this syndrome. By using a hybrid facial-recognition model, we show that individuals with a pathogenic variant in KDM3B have a facial gestalt, and that they show significant facial similarity compared to control individuals with ID. In conclusion, pathogenic variants in KDM3B cause a syndrome characterized by ID, short stature, and facial dysmorphism.
- Published
- 2019
30. Outcomes of patients with childhood B-cell precursor acute lymphoblastic leukaemia with late bone marrow relapses: long-term follow-up of the ALLR3 open-label randomised trial
- Author
-
Parker, C., Krishnan, S., Hamadeh, L., Irving, J.A., Kuiper, R.P., Revesz, T., Hoogerbrugge, P.M., Hancock, J., Sutton, R., Moorman, A.V., Saha, V., Parker, C., Krishnan, S., Hamadeh, L., Irving, J.A., Kuiper, R.P., Revesz, T., Hoogerbrugge, P.M., Hancock, J., Sutton, R., Moorman, A.V., and Saha, V.
- Abstract
Contains fulltext : 207593.pdf (publisher's version ) (Open Access), BACKGROUND: The ALLR3 trial investigated outcomes of children with B-cell precursor acute lymphoblastic leukaemia who had late bone marrow relapses. We analysed long-term follow-up outcomes of these patients. METHODS: ALLR3 was an open-label randomised clinical trial that recruited children aged 1-18 years with B-cell precursor acute lymphoblastic leukaemia who had late bone marrow relapses. Eligible patients were recruited from centres in Australia, Ireland, the Netherlands, New Zealand, and the UK. Patients were randomly assigned from Jan 31, 2003, to Dec 31, 2007, and the trial closed to recruitment on Oct 31, 2013. Randomly assigned patients were allocated to receive either idarubicin or mitoxantrone in induction by stratified concealed randomisation; after randomisation stopped in Dec 31, 2007, all patients were allocated to receive mitoxantrone. After three blocks of therapy, patients with high minimal residual disease (>/=10(-4) cells) at the end of induction were allocated to undergo allogeneic stem-cell transplantation and those with low minimal residual disease (<10(-4) cells) at the end of induction were allocated to receive chemotherapy. Minimal residual disease level was measured by real-time quantitative PCR analysis of immunoglobulin and T-cell receptor gene rearrangements. The primary endpoint of the original ALLR3 clinical trial was progression-free survival of randomly assigned patients. The primary endpoint of this long-term follow-up analysis was progression-free survival of patients with late bone marrow relapses stratified by minimal residual disease level. Outcomes were correlated with age, site, time to recurrence, and genetic subtypes, and analysed by both intention to treat and actual treatment received. This trial is registered on the ISRCTN registry, number ISRCTN45724312, and on ClinicalTrials.gov, number NCT00967057. FINDINGS: Between Feb 2, 2003, and Oct 28, 2013, 228 patients with B-cell precursor acute lymphoblastic leukaemia and lat
- Published
- 2019
31. Mutational Signature Analysis Reveals NTHL1 Deficiency to Cause a Multi-tumor Phenotype
- Author
-
Grolleman, J.E., Voer, R.M. de, Elsayed, F.A., Nielsen, Maartje, Weren, R.D.A., Palles, Claire, Ligtenberg, M.J.L., Vos, J.R., Kuiper, R.A., Kamping, E.J., Jansen, E, Jongmans, M.C.J., Neveling, K., Geurts van Kessel, A., Hoogerbrugge, N., Kuiper, R.P., Grolleman, J.E., Voer, R.M. de, Elsayed, F.A., Nielsen, Maartje, Weren, R.D.A., Palles, Claire, Ligtenberg, M.J.L., Vos, J.R., Kuiper, R.A., Kamping, E.J., Jansen, E, Jongmans, M.C.J., Neveling, K., Geurts van Kessel, A., Hoogerbrugge, N., and Kuiper, R.P.
- Abstract
Contains fulltext : 201344.pdf (publisher's version ) (Closed access)
- Published
- 2019
32. High sensitivity and clonal stability of the genomic fusion as single marker for response monitoring in ETV6-RUNX1-positive acute lymphoblastic leukemia
- Author
-
Hoffmann, J., Krumbholz, M., Gutierrez, H.P., Fillies, M., Szymansky, A., Bleckmann, K., Stadt, U. Zur, Kohler, R., Kuiper, R.P., Horstmann, M., Stackelberg, A. von, Eckert, C., Metzler, M., Hoffmann, J., Krumbholz, M., Gutierrez, H.P., Fillies, M., Szymansky, A., Bleckmann, K., Stadt, U. Zur, Kohler, R., Kuiper, R.P., Horstmann, M., Stackelberg, A. von, Eckert, C., and Metzler, M.
- Abstract
Item does not contain fulltext, BACKGROUND: Assessment of minimal residual disease (MRD) is an integral component for response monitoring and treatment stratification in acute lymphoblastic leukemia (ALL). We aimed to evaluate the genomic ETV6-RUNX1 fusion sites as a single marker for MRD quantification. PROCEDURE: In a representative, uniformly treated cohort of pediatric relapsed ALL patients (n = 52), ETV6-RUNX1 fusion sites were compared to the current gold standard, immunoglobulin/T-cell receptor (Ig/TCR) gene rearrangements. RESULTS: Primer/probe sets designed to ETV6-RUNX1 fusions achieved significantly more frequent a sensitivity and a quantitative range of at least 10(-4) compared to the gold standard with 100% and 73% versus 76% and 47%, respectively. The breakpoint sequence was identical at diagnosis and relapse in all tested cases. There was a high degree of concordance between quantitative MRD results assessed using ETV6-RUNX1 and the highest Ig/TCR marker (Spearman's 0.899, P < .01) with differences >(1/2) log-step in only 6% of patients. A high proportion of ETV6-RUNX1-positive ALL relapses (40%) in our cohort showed a poor response to induction treatment at relapse, and therefore had an indication for hematopoietic stem cell transplantation, demonstrating the need of accurate identification of this subgroup. CONCLUSIONS: ETV6-RUNX1 fusion sites are highly sensitive and reliable MRD markers. Our data confirm that they are unaffected by clonal evolution and selection during front-line and second-line chemotherapy in contrast to Ig/TCR rearrangements, which require several markers per patient to compensate for the observed loss of target clones. In future studies, the genomic ETV6-RUNX1 fusion can be used as single MRD marker.
- Published
- 2019
33. TRIM28 haploinsufficiency predisposes to Wilms tumor
- Author
-
Diets, I.J., Hoyer, J., Ekici, A.B., Popp, B., Hoogerbrugge, N., Reijmersdal, S.V. van, Bhaskaran, R., Hadjihannas, M., Vasileiou, G., Thiel, C.T., Seven, D., Uebe, S., Ilencikova, D., Waanders, E., Mavinkurve-Groothuis, A.M.C., Roeleveld, N., Krijger, R.R. de, Wegert, J., Graf, N., Vokuhl, C., Agaimy, A., Gessler, M., Reis, A., Kuiper, R.P., Jongmans, M.C.J., Metzler, M., Diets, I.J., Hoyer, J., Ekici, A.B., Popp, B., Hoogerbrugge, N., Reijmersdal, S.V. van, Bhaskaran, R., Hadjihannas, M., Vasileiou, G., Thiel, C.T., Seven, D., Uebe, S., Ilencikova, D., Waanders, E., Mavinkurve-Groothuis, A.M.C., Roeleveld, N., Krijger, R.R. de, Wegert, J., Graf, N., Vokuhl, C., Agaimy, A., Gessler, M., Reis, A., Kuiper, R.P., Jongmans, M.C.J., and Metzler, M.
- Abstract
Contains fulltext : 205528.pdf (publisher's version ) (Closed access), Two percent of patients with Wilms tumors have a positive family history. In many of these cases the genetic cause remains unresolved. By applying germline exome sequencing in two families with two affected individuals with Wilms tumors, we identified truncating mutations in TRIM28. Subsequent mutational screening of germline and tumor DNA of 269 children affected by Wilms tumor was performed, and revealed seven additional individuals with germline truncating mutations, and one individual with a somatic truncating mutation in TRIM28. TRIM28 encodes a complex scaffold protein involved in many different processes, including gene silencing, DNA repair and maintenance of genomic integrity. Expression studies on mRNA and protein level showed reduction of TRIM28, confirming a loss-of-function effect of the mutations identified. The tumors showed an epithelial-type histology that stained negative for TRIM28 by immunohistochemistry. The tumors were bilateral in six patients, and 10/11 tumors are accompanied by perilobar nephrogenic rests. Exome sequencing on eight tumor DNA samples from six individuals showed loss-of-heterozygosity (LOH) of the TRIM28-locus by mitotic recombination in seven tumors, suggesting that TRIM28 functions as a tumor suppressor gene in Wilms tumor development. Additionally, the tumors showed very few mutations in known Wilms tumor driver genes, suggesting that loss of TRIM28 is the main driver of tumorigenesis. In conclusion, we identified heterozygous germline truncating mutations in TRIM28 in 11 children with mainly epithelial-type Wilms tumors, which become homozygous in tumor tissue. These data establish TRIM28 as a novel Wilms tumor predisposition gene, acting as a tumor suppressor gene by LOH.
- Published
- 2019
34. Validation of the United Kingdom copy-number alteration classifier in 3239 children with B-cell precursor ALL
- Author
-
Hamadeh, L., Enshaei, A., Schwab, C., Alonso, C.N., Attarbaschi, A., Barbany, G., Boer, M.L. Den, Boer, J.M.A., Braun, M., Pozza, L. Dalla, Elitzur, S., Emerenciano, M., Fechina, L., Felice, M.S., Fronkova, E., Haltrich, I., Heyman, M.M., Horibe, K., Imamura, T., Jeison, M., Kovacs, G., Kuiper, R.P., Mlynarski, W., Nebral, K., Ofverholm, I. Ivanov, Pastorczak, A., Pieters, R., Piko, H., Pombo-de-Oliveira, M.S., Rubio, P., Strehl, S., Stary, J., Sutton, R., Trka, J., Tsaur, G., Venn, N., Vora, A., Yano, M., Harrison, C.J., Moorman, A.V., Hamadeh, L., Enshaei, A., Schwab, C., Alonso, C.N., Attarbaschi, A., Barbany, G., Boer, M.L. Den, Boer, J.M.A., Braun, M., Pozza, L. Dalla, Elitzur, S., Emerenciano, M., Fechina, L., Felice, M.S., Fronkova, E., Haltrich, I., Heyman, M.M., Horibe, K., Imamura, T., Jeison, M., Kovacs, G., Kuiper, R.P., Mlynarski, W., Nebral, K., Ofverholm, I. Ivanov, Pastorczak, A., Pieters, R., Piko, H., Pombo-de-Oliveira, M.S., Rubio, P., Strehl, S., Stary, J., Sutton, R., Trka, J., Tsaur, G., Venn, N., Vora, A., Yano, M., Harrison, C.J., and Moorman, A.V.
- Abstract
Contains fulltext : 204641.pdf (publisher's version ) (Open Access), Genetic abnormalities provide vital diagnostic and prognostic information in pediatric acute lymphoblastic leukemia (ALL) and are increasingly used to assign patients to risk groups. We recently proposed a novel classifier based on the copy-number alteration (CNA) profile of the 8 most commonly deleted genes in B-cell precursor ALL. This classifier defined 3 CNA subgroups in consecutive UK trials and was able to discriminate patients with intermediate-risk cytogenetics. In this study, we sought to validate the United Kingdom ALL (UKALL)-CNA classifier and reevaluate the interaction with cytogenetic risk groups using individual patient data from 3239 cases collected from 12 groups within the International BFM Study Group. The classifier was validated and defined 3 risk groups with distinct event-free survival (EFS) rates: good (88%), intermediate (76%), and poor (68%) (P < .001). There was no evidence of heterogeneity, even within trials that used minimal residual disease to guide therapy. By integrating CNA and cytogenetic data, we replicated our original key observation that patients with intermediate-risk cytogenetics can be stratified into 2 prognostic subgroups. Group A had an EFS rate of 86% (similar to patients with good-risk cytogenetics), while group B patients had a significantly inferior rate (73%, P < .001). Finally, we revised the overall genetic classification by defining 4 risk groups with distinct EFS rates: very good (91%), good (81%), intermediate (73%), and poor (54%), P < .001. In conclusion, the UKALL-CNA classifier is a robust prognostic tool that can be deployed in different trial settings and used to refine established cytogenetic risk groups.
- Published
- 2019
35. Accurate detection of low-level mosaic mutations in pediatric acute lymphoblastic leukemia using single molecule tagging and deep-sequencing
- Author
-
Yu, J., Antic, Zeljko, Reijmersdal, S.V. van, Hoischen, A., Sonneveld, E., Waanders, E., Kuiper, R.P., Yu, J., Antic, Zeljko, Reijmersdal, S.V. van, Hoischen, A., Sonneveld, E., Waanders, E., and Kuiper, R.P.
- Abstract
Item does not contain fulltext
- Published
- 2018
36. High Yield of Pathogenic Germline Mutations Causative or Likely Causative of the Cancer Phenotype in Selected Children with Cancer
- Author
-
Diets, I.J., Waanders, E., Ligtenberg, M.J.L., Bladel, D.A.G. van, Kamping, E.J., Hoogerbrugge, P.M., Hopman, S., Olderode-Berends, M.J., Gerkes, E.H., Koolen, D.A., Marcelis, C.L., Santen, G.W.E., Belzen, M.J. van, Mordaunt, D., McGregor, L., Thompson, E., Kattamis, A., Pastorczak, A., Mlynarski, W., Ilencikova, D., Vulto-van Silfhout, A.T., Gardeitchik, T., Bont, E.S. de, Loeffen, J., Wagner, A., Mensenkamp, A.R., Kuiper, R.P., Hoogerbrugge, N., Jongmans, M.C., Diets, I.J., Waanders, E., Ligtenberg, M.J.L., Bladel, D.A.G. van, Kamping, E.J., Hoogerbrugge, P.M., Hopman, S., Olderode-Berends, M.J., Gerkes, E.H., Koolen, D.A., Marcelis, C.L., Santen, G.W.E., Belzen, M.J. van, Mordaunt, D., McGregor, L., Thompson, E., Kattamis, A., Pastorczak, A., Mlynarski, W., Ilencikova, D., Vulto-van Silfhout, A.T., Gardeitchik, T., Bont, E.S. de, Loeffen, J., Wagner, A., Mensenkamp, A.R., Kuiper, R.P., Hoogerbrugge, N., and Jongmans, M.C.
- Abstract
Contains fulltext : 190442.pdf (publisher's version ) (Closed access), Purpose: In many children with cancer and characteristics suggestive of a genetic predisposition syndrome, the genetic cause is still unknown. We studied the yield of pathogenic mutations by applying whole-exome sequencing on a selected cohort of children with cancer.Experimental Design: To identify mutations in known and novel cancer-predisposing genes, we performed trio-based whole-exome sequencing on germline DNA of 40 selected children and their parents. These children were diagnosed with cancer and had at least one of the following features: (1) intellectual disability and/or congenital anomalies, (2) multiple malignancies, (3) family history of cancer, or (4) an adult type of cancer. We first analyzed the sequence data for germline mutations in 146 known cancer-predisposing genes. If no causative mutation was found, the analysis was extended to the whole exome.Results: Four patients carried causative mutations in a known cancer-predisposing gene: TP53 and DICER1 (n = 3). In another 4 patients, exome sequencing revealed mutations causing syndromes that might have contributed to the malignancy (EP300-based Rubinstein-Taybi syndrome, ARID1A-based Coffin-Siris syndrome, ACTB-based Baraitser-Winter syndrome, and EZH2-based Weaver syndrome). In addition, we identified two genes, KDM3B and TYK2, which are possibly involved in genetic cancer predisposition.Conclusions: In our selected cohort of patients, pathogenic germline mutations causative or likely causative of the cancer phenotype were found in 8 patients, and two possible novel cancer-predisposing genes were identified. Therewith, our study shows the added value of sequencing beyond a cancer gene panel in selected patients, to recognize childhood cancer predisposition. Clin Cancer Res; 24(7); 1594-603. (c)2018 AACR.
- Published
- 2018
37. Tumor suppressors BTG1 and IKZF1 cooperate during mouse leukemia development and increase relapse risk in B-cell precursor acute lymphoblastic leukemia patients
- Author
-
Scheijen, B., Boer, J.M.A., Marke, R., Tijchon, E.J., Ingen Schenau, D.S. van, Waanders, E., Emst, L. van, Meer, L.T. van der, Pieters, R., Escherich, G., Horstmann, M.A., Sonneveld, E., Venn, N., Sutton, R., Dalla-Pozza, L., Kuiper, R.P., Hoogerbrugge, P.M., Boer, M.L. Den, Leeuwen, F.N. van, Scheijen, B., Boer, J.M.A., Marke, R., Tijchon, E.J., Ingen Schenau, D.S. van, Waanders, E., Emst, L. van, Meer, L.T. van der, Pieters, R., Escherich, G., Horstmann, M.A., Sonneveld, E., Venn, N., Sutton, R., Dalla-Pozza, L., Kuiper, R.P., Hoogerbrugge, P.M., Boer, M.L. Den, and Leeuwen, F.N. van
- Abstract
Contains fulltext : 169866.pdf (publisher's version ) (Open Access), Deletions and mutations affecting lymphoid transcription factor IKZF1 (IKAROS) are associated with an increased relapse risk and poor outcome in B-cell precursor acute lymphoblastic leukemia. However, additional genetic events may either enhance or negate the effects of IKZF1 deletions on prognosis. In a large discovery cohort of 533 childhood B-cell precursor acute lymphoblastic leukemia patients, we observed that single-copy losses of BTG1 were significantly enriched in IKZF1-deleted B-cell precursor acute lymphoblastic leukemia (P=0.007). While BTG1 deletions alone had no impact on prognosis, the combined presence of BTG1 and IKZF1 deletions was associated with a significantly lower 5-year event-free survival (P=0.0003) and a higher 5-year cumulative incidence of relapse (P=0.005), when compared with IKZF1-deleted cases without BTG1 aberrations. In contrast, other copy number losses commonly observed in B-cell precursor acute lymphoblastic leukemia, such as CDKN2A/B, PAX5, EBF1 or RB1, did not affect the outcome of IKZF1-deleted acute lymphoblastic leukemia patients. To establish whether the combined loss of IKZF1 and BTG1 function cooperate in leukemogenesis, Btg1-deficient mice were crossed onto an Ikzf1 heterozygous background. We observed that loss of Btg1 increased the tumor incidence of Ikzf1+/- mice in a dose-dependent manner. Moreover, murine B cells deficient for Btg1 and Ikzf1+/- displayed increased resistance to glucocorticoids, but not to other chemotherapeutic drugs. Together, our results identify BTG1 as a tumor suppressor in leukemia that, when deleted, strongly enhances the risk of relapse in IKZF1-deleted B-cell precursor acute lymphoblastic leukemia, and augments the glucocorticoid resistance phenotype mediated by the loss of IKZF1 function.
- Published
- 2017
38. Surveillance Recommendations for Children with Overgrowth Syndromes and Predisposition to Wilms Tumors and Hepatoblastoma
- Author
-
Kalish, J.M., Doros, L., Helman, L.J., Hennekam, R.C., Kuiper, R.P., Maas, S.M., Maher, E.R., Nichols, K.E., Plon, S.E., Porter, C.C., Rednam, S., Schultz, K.A.P., States, L.J., Tomlinson, G.E., Zelley, K., Druley, T.E., Kalish, J.M., Doros, L., Helman, L.J., Hennekam, R.C., Kuiper, R.P., Maas, S.M., Maher, E.R., Nichols, K.E., Plon, S.E., Porter, C.C., Rednam, S., Schultz, K.A.P., States, L.J., Tomlinson, G.E., Zelley, K., and Druley, T.E.
- Abstract
Item does not contain fulltext, A number of genetic syndromes have been linked to increased risk for Wilms tumor (WT), hepatoblastoma (HB), and other embryonal tumors. Here, we outline these rare syndromes with at least a 1% risk to develop these tumors and recommend uniform tumor screening recommendations for North America. Specifically, for syndromes with increased risk for WT, we recommend renal ultrasounds every 3 months from birth (or the time of diagnosis) through the seventh birthday. For HB, we recommend screening with full abdominal ultrasound and alpha-fetoprotein serum measurements every 3 months from birth (or the time of diagnosis) through the fourth birthday. We recommend that when possible, these patients be evaluated and monitored by cancer predisposition specialists. At this time, these recommendations are not based on the differential risk between different genetic or epigenetic causes for each syndrome, which some European centers have implemented. This differentiated approach largely represents distinct practice environments between the United States and Europe, and these guidelines are designed to be a broad framework within which physicians and families can work together to implement specific screening. Further study is expected to lead to modifications of these recommendations. Clin Cancer Res; 23(13); e115-e22. (c)2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.
- Published
- 2017
39. Tyrosine kinase fusion genes in pediatric BCR-ABL1-like acute lymphoblastic leukemia
- Author
-
Boer, J.M.A., Steeghs, E.M.P., Marchante, J.R., Boeree, A., Beaudoin, J.J., Beverloo, H.B., Kuiper, R.P., Escherich, G., Velden, V.H. van der, Schoot, C.E. van der, Groot-Kruseman, H.A. de, Pieters, R., Boer, M.L. Den, Boer, J.M.A., Steeghs, E.M.P., Marchante, J.R., Boeree, A., Beaudoin, J.J., Beverloo, H.B., Kuiper, R.P., Escherich, G., Velden, V.H. van der, Schoot, C.E. van der, Groot-Kruseman, H.A. de, Pieters, R., and Boer, M.L. Den
- Abstract
Contains fulltext : 169868.pdf (Publisher’s version ) (Open Access), Approximately 15% of pediatric B cell precursor acute lymphoblastic leukemia (BCP-ALL) is characterized by gene expression similar to that of BCR-ABL1-positive disease and unfavorable prognosis. This BCR-ABL1-like subtype shows a high frequency of B-cell development gene aberrations and tyrosine kinase-activating lesions. To evaluate the clinical significance of tyrosine kinase gene fusions in children with BCP-ALL, we studied the frequency of recently identified tyrosine kinase fusions, associated genetic features, and prognosis in a representative Dutch/German cohort. We identified 14 tyrosine kinase fusions among 77 BCR-ABL1-like cases (18%) and none among 76 non-BCR-ABL1-like B-other cases. Novel exon fusions were identified for RCSD1-ABL2 and TERF2-JAK2. JAK2 mutation was mutually exclusive with tyrosine kinase fusions and only occurred in cases with high CRLF2 expression. The non/late response rate and levels of minimal residual disease in the fusion-positive BCR-ABL1-like group were higher than in the non-BCR-ABL1-like B-others (p<0.01), and also higher, albeit not statistically significant, compared with the fusion-negative BCR-ABL1-like group. The 8-year cumulative incidence of relapse in the fusion-positive BCR-ABL1-like group (35%) was comparable with that in the fusion-negative BCR-ABL1-like group (35%), and worse than in the non-BCR-ABL1-like B-other group (17%, p=0.07). IKZF1 deletions, predominantly other than the dominant-negative isoform and full deletion, co-occurred with tyrosine kinase fusions. This study shows that tyrosine kinase fusion-positive cases are a high-risk subtype of BCP-ALL, which warrants further studies with specific kinase inhibitors to improve outcome.
- Published
- 2017
40. Unraveling genetic predisposition to familial or early onset gastric cancer using germline whole-exome sequencing
- Author
-
Vogelaar, I.P., Post, R.S. van der, Krieken, J.H. van, Spruijt, L., Zelst-Stams, W.A.G. van, Kets, C.M., Lubinski, J., Jakubowska, A., Teodorczyk, U., Aalfs, C.M., Hest, L.P. van, Pinheiro, H., Oliveira, C. de, Jhangiani, S.N., Muzny, D.M., Gibbs, R.A., Lupski, J.R., Ligt, J. de, Vissers, L.E.L.M., Hoischen, A., Gilissen, C., Vorst, J.M. van de, Goeman, J.J., Schackert, H.K., Ranzani, G.N., Molinaro, V., Garcia, E.B., Hes, F.J., Holinski-Feder, E., Genuardi, M., Ausems, M., Sijmons, R.H., Wagner, A., Kolk, L.E. van der, Bjornevoll, I., Hoberg-Vetti, H., Geurts van Kessel, A.H.M., Kuiper, R.P., Ligtenberg, M.J.L., Hoogerbrugge, N., Vogelaar, I.P., Post, R.S. van der, Krieken, J.H. van, Spruijt, L., Zelst-Stams, W.A.G. van, Kets, C.M., Lubinski, J., Jakubowska, A., Teodorczyk, U., Aalfs, C.M., Hest, L.P. van, Pinheiro, H., Oliveira, C. de, Jhangiani, S.N., Muzny, D.M., Gibbs, R.A., Lupski, J.R., Ligt, J. de, Vissers, L.E.L.M., Hoischen, A., Gilissen, C., Vorst, J.M. van de, Goeman, J.J., Schackert, H.K., Ranzani, G.N., Molinaro, V., Garcia, E.B., Hes, F.J., Holinski-Feder, E., Genuardi, M., Ausems, M., Sijmons, R.H., Wagner, A., Kolk, L.E. van der, Bjornevoll, I., Hoberg-Vetti, H., Geurts van Kessel, A.H.M., Kuiper, R.P., Ligtenberg, M.J.L., and Hoogerbrugge, N.
- Abstract
Contains fulltext : 182216.pdf (publisher's version ) (Open Access), Recognition of individuals with a genetic predisposition to gastric cancer (GC) enables preventive measures. However, the underlying cause of genetic susceptibility to gastric cancer remains largely unexplained. We performed germline whole-exome sequencing on leukocyte DNA of 54 patients from 53 families with genetically unexplained diffuse-type and intestinal-type GC to identify novel GC-predisposing candidate genes. As young age at diagnosis and familial clustering are hallmarks of genetic tumor susceptibility, we selected patients that were diagnosed below the age of 35, patients from families with two cases of GC at or below age 60 and patients from families with three GC cases at or below age 70. All included individuals were tested negative for germline CDH1 mutations before or during the study. Variants that were possibly deleterious according to in silico predictions were filtered using several independent approaches that were based on gene function and gene mutation burden in controls. Despite a rigorous search, no obvious candidate GC predisposition genes were identified. This negative result stresses the importance of future research studies in large, homogeneous cohorts.
- Published
- 2017
41. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer
- Author
-
Drost, Jarno, Boxtel, R. van, Blokzijl, F., Mizutani, T., Sasaki, N., Sasselli, V., Ligt, J. de, Behjati, S., Grolleman, J.E., Wezel, T. van, Nik-Zainal, S., Kuiper, R.P., Cuppen, E., Clevers, H., Drost, Jarno, Boxtel, R. van, Blokzijl, F., Mizutani, T., Sasaki, N., Sasselli, V., Ligt, J. de, Behjati, S., Grolleman, J.E., Wezel, T. van, Nik-Zainal, S., Kuiper, R.P., Cuppen, E., and Clevers, H.
- Abstract
Item does not contain fulltext, Mutational processes underlie cancer initiation and progression. Signatures of these processes in cancer genomes may explain cancer etiology and could hold diagnostic and prognostic value. We developed a strategy that can be used to explore the origin of cancer-associated mutational signatures. We used CRISPR-Cas9 technology to delete key DNA repair genes in human colon organoids, followed by delayed subcloning and whole-genome sequencing. We found that mutation accumulation in organoids deficient in the mismatch repair gene MLH1 is driven by replication errors and accurately models the mutation profiles observed in mismatch repair-deficient colorectal cancers. Application of this strategy to the cancer predisposition gene NTHL1, which encodes a base excision repair protein, revealed a mutational footprint (signature 30) previously observed in a breast cancer cohort. We show that signature 30 can arise from germline NTHL1 mutations.
- Published
- 2017
42. Cancer Screening Recommendations and Clinical Management of Inherited Gastrointestinal Cancer Syndromes in Childhood
- Author
-
Achatz, M.I., Porter, C.C., Brugieres, L., Druker, H., Frebourg, T., Foulkes, W.D., Kratz, C.P., Kuiper, R.P., Hansford, J.R., Hernandez, H.S., Nathanson, K.L., Kohlmann, W.K., Doros, L., Onel, K., Schneider, K.W, Scollon, S.R., Tabori, U., Tomlinson, G.E., Evans, D.G., Plon, S.E., Achatz, M.I., Porter, C.C., Brugieres, L., Druker, H., Frebourg, T., Foulkes, W.D., Kratz, C.P., Kuiper, R.P., Hansford, J.R., Hernandez, H.S., Nathanson, K.L., Kohlmann, W.K., Doros, L., Onel, K., Schneider, K.W, Scollon, S.R., Tabori, U., Tomlinson, G.E., Evans, D.G., and Plon, S.E.
- Abstract
Item does not contain fulltext, Hereditary gastrointestinal cancer predisposition syndromes have been well characterized, but management strategies and surveillance remain a major challenge, especially in childhood. In October 2016, the American Association for Cancer Research organized the AACR Childhood Cancer Predisposition Workshop in which international experts in care of children with a hereditary risk of cancer met to define surveillance strategies and management of children with cancer predisposition syndromes. In this article, we review the current literature in polyposis syndromes that can be diagnosed in childhood and may be associated with an increased incidence of gastrointestinal neoplasms and other cancer types. These disorders include adenomatous polyposis syndromes (APC and MUTYH), juvenile polyposis coli (BMPR1A and SMAD4), Peutz-Jeghers Syndrome (STK11/LKB1), and PTEN hamartoma tumor syndrome (PHTS; PTEN), which can present with a more limited juvenile polyposis phenotype. Herein, the panel of experts provides recommendations for clinical diagnosis, approach to genetic testing, and focus on cancer surveillance recommendations when appropriate during the pediatric period. We also review current controversies on genetic evaluation of patients with hepatoblastoma and indications for surveillance for this tumor. Childhood cancer risks and surveillance associated with disorders involving the mismatch repair genes, including Lynch syndrome and constitutional mismatch repair deficiency (CMMRD), are discussed elsewhere in this series. Clin Cancer Res; 23(13); e107-e14. (c)2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.
- Published
- 2017
43. Antagonism of B cell enhancer networks by STAT5 drives leukemia and poor patient survival
- Author
-
Katerndahl, C.D.S., Heltemes-Harris, L.M., Willette, M.J.L., Henzler, C.M., Frietze, S., Yang, R., Schjerven, H., Silverstein, K.A.T., Ramsey, L.B., Hubbard, G., Wells, A.D., Kuiper, R.P, Scheijen, B., Leeuwen, F.N. van, Muschen, M., Kornblau, S.M., Farrar, M.A., Katerndahl, C.D.S., Heltemes-Harris, L.M., Willette, M.J.L., Henzler, C.M., Frietze, S., Yang, R., Schjerven, H., Silverstein, K.A.T., Ramsey, L.B., Hubbard, G., Wells, A.D., Kuiper, R.P, Scheijen, B., Leeuwen, F.N. van, Muschen, M., Kornblau, S.M., and Farrar, M.A.
- Abstract
Item does not contain fulltext, The transcription factor STAT5 has a critical role in B cell acute lymphoblastic leukemia (B-ALL). How STAT5 mediates this effect is unclear. Here we found that activation of STAT5 worked together with defects in signaling components of the precursor to the B cell antigen receptor (pre-BCR), including defects in BLNK, BTK, PKCbeta, NF-kappaB1 and IKAROS, to initiate B-ALL. STAT5 antagonized the transcription factors NF-kappaB and IKAROS by opposing regulation of shared target genes. Super-enhancers showed enrichment for STAT5 binding and were associated with an opposing network of transcription factors, including PAX5, EBF1, PU.1, IRF4 and IKAROS. Patients with a high ratio of active STAT5 to NF-kappaB or IKAROS had more-aggressive disease. Our studies indicate that an imbalance of two opposing transcriptional programs drives B-ALL and suggest that restoring the balance of these pathways might inhibit B-ALL.
- Published
- 2017
44. Recommendations for Surveillance for Children with Leukemia-Predisposing Conditions
- Author
-
Porter, C.C., Druley, T.E., Erez, A., Kuiper, R.P., Onel, K., Schiffman, J.D., Schneider, K.W, Scollon, S.R., Scott, H.S., Strong, L.C., Walsh, M.F., Nichols, K.E., Porter, C.C., Druley, T.E., Erez, A., Kuiper, R.P., Onel, K., Schiffman, J.D., Schneider, K.W, Scollon, S.R., Scott, H.S., Strong, L.C., Walsh, M.F., and Nichols, K.E.
- Abstract
Item does not contain fulltext, Leukemia, the most common childhood cancer, has long been recognized to occasionally run in families. The first clues about the genetic mechanisms underlying familial leukemia emerged in 1990 when Li-Fraumeni syndrome was linked to TP53 mutations. Since this discovery, many other genes associated with hereditary predisposition to leukemia have been identified. Although several of these disorders also predispose individuals to solid tumors, certain conditions exist in which individuals are specifically at increased risk to develop myelodysplastic syndrome (MDS) and/or acute leukemia. The increasing identification of affected individuals and families has raised questions around the efficacy, timing, and optimal methods of surveillance. As part of the AACR Childhood Cancer Predisposition Workshop, an expert panel met to review the spectrum of leukemia-predisposing conditions, with the aim to develop consensus recommendations for surveillance for pediatric patients. The panel recognized that for several conditions, routine monitoring with complete blood counts and bone marrow evaluations is essential to identify disease evolution and enable early intervention with allogeneic hematopoietic stem cell transplantation. However, for others, less intensive surveillance may be considered. Because few reports describing the efficacy of surveillance exist, the recommendations derived by this panel are based on opinion, and local experience and will need to be revised over time. The development of registries and clinical trials is urgently needed to enhance understanding of the natural history of the leukemia-predisposing conditions, such that these surveillance recommendations can be optimized to further enhance long-term outcomes. Clin Cancer Res; 23(11); e14-e22. (c)2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.
- Published
- 2017
45. A molecular inversion probe-based next-generation sequencing panel to detect germline mutations in Chinese early-onset colorectal cancer patients.
- Author
-
Zhang, J., Wang, W., Voer, R.M. de, Hehir-Kwa, J.Y., Kamping, E.J., Weren, R.D.A., Nelen, M.R., Hoischen, A., Ligtenberg, M.J.L., Hoogerbrugge, N., Yang, X, Yang, Z, Fan, X., Wang, L., Liu, H., Wang, J, Kuiper, R.P., Geurts van Kessel, A.H.M., Zhang, J., Wang, W., Voer, R.M. de, Hehir-Kwa, J.Y., Kamping, E.J., Weren, R.D.A., Nelen, M.R., Hoischen, A., Ligtenberg, M.J.L., Hoogerbrugge, N., Yang, X, Yang, Z, Fan, X., Wang, L., Liu, H., Wang, J, Kuiper, R.P., and Geurts van Kessel, A.H.M.
- Abstract
Contains fulltext : 169654.pdf (publisher's version ) (Open Access)
- Published
- 2017
46. Germline activating TYK2 mutations in pediatric patients with two primary acute lymphoblastic leukemia occurrences
- Author
-
Waanders, E., Scheijen, B., Jongmans, M.C.J., Venselaar, H., Reijmersdal, S.V. van, Dijk, A.H.A. van, Pastorczak, A., Weren, R.D.A., Schoot, C.E. van der, Vorst, J.M. van de, Sonneveld, E., Hoogerbrugge, N., Velden, V.H. van der, Gruhn, B., Hoogerbrugge, P.M., Dongen, J.J. van, Geurts van Kessel, A.H.M., Leeuwen, F.N. van, Kuiper, R.P., Waanders, E., Scheijen, B., Jongmans, M.C.J., Venselaar, H., Reijmersdal, S.V. van, Dijk, A.H.A. van, Pastorczak, A., Weren, R.D.A., Schoot, C.E. van der, Vorst, J.M. van de, Sonneveld, E., Hoogerbrugge, N., Velden, V.H. van der, Gruhn, B., Hoogerbrugge, P.M., Dongen, J.J. van, Geurts van Kessel, A.H.M., Leeuwen, F.N. van, and Kuiper, R.P.
- Abstract
Contains fulltext : 170242.pdf (publisher's version ) (Closed access)
- Published
- 2017
47. Prevalence of germline mutations in the spindle assembly checkpoint gene BUB1B in individuals with early-onset colorectal cancer
- Author
-
Hahn, M.M., Vreede, L.A., Bemelmans, S.AS.A., Looij, E. van der, Geurts van Kessel, A.H.M., Schackert, H.K., Ligtenberg, M.J., Hoogerbrugge, N., Kuiper, R.P., and Voer, R.M. de
- Subjects
Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] - Abstract
Item does not contain fulltext Germline mutations in BUB1B, encoding BUBR1, one of the crucial components of the spindle assembly checkpoint (SAC), have been shown to cause variable phenotypes, including the recessive mosaic variegated aneuploidy (MVA) syndrome, which predisposes to cancer. Reduced levels of the wild-type BUBR1 protein have been linked to the development of gastrointestinal neoplasms. To determine whether mutations in BUB1B are enriched in individuals with colorectal cancer (CRC), we performed amplicon-based targeted next-generation sequencing of BUB1B on germline DNA of 192 individuals with early-onset CRC (
- Published
- 2016
48. Nieuwe genomische markers voor diagnostiek en behandeling van voorloper-b-cel acute lymfatische leukemie bij kinderen
- Author
-
Kuiper, R.P., Hoogerbrugge, P.M., and Leeuwen, F.N. van
- Subjects
GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,Age-related aspects of cancer Immune Regulation [ONCOL 2] - Abstract
Contains fulltext : 89729.pdf (Publisher’s version ) (Open Access)
- Published
- 2010
49. Tumor suppressor IKZF1 mediates glucocorticoid resistance in B-cell precursor acute lymphoblastic leukemia
- Author
-
Marke, R., Havinga, J., Cloos, J., Demkes, M., Poelmans, G., Yuniati, L., Ingen Schenau, D.S. van, Sonneveld, E., Waanders, E., Pieters, R., Kuiper, R.P., Hoogerbrugge, P.M., Kaspers, G.J., Leeuwen, F.N. van, Scheijen, B., Marke, R., Havinga, J., Cloos, J., Demkes, M., Poelmans, G., Yuniati, L., Ingen Schenau, D.S. van, Sonneveld, E., Waanders, E., Pieters, R., Kuiper, R.P., Hoogerbrugge, P.M., Kaspers, G.J., Leeuwen, F.N. van, and Scheijen, B.
- Abstract
Item does not contain fulltext
- Published
- 2016
50. Analyzing structure-function relationships of artificial and cancer-associated PARP1 variants by reconstituting TALEN-generated HeLa PARP1 knock-out cells
- Author
-
Rank, L., Veith, S., Gwosch, E.C., Demgenski, J., Ganz, M., Jongmans, M.C.J., Vogel, C., Fischbach, A., Buerger, S., Fischer, J.M., Zubel, T., Stier, A., Renner, C., Schmalz, M., Beneke, S., Groettrup, M., Kuiper, R.P., Burkle, A., Ferrando-May, E., Mangerich, A., Rank, L., Veith, S., Gwosch, E.C., Demgenski, J., Ganz, M., Jongmans, M.C.J., Vogel, C., Fischbach, A., Buerger, S., Fischer, J.M., Zubel, T., Stier, A., Renner, C., Schmalz, M., Beneke, S., Groettrup, M., Kuiper, R.P., Burkle, A., Ferrando-May, E., and Mangerich, A.
- Abstract
Contains fulltext : 171327.pdf (publisher's version ) (Open Access), Genotoxic stress activates PARP1, resulting in the post-translational modification of proteins with poly(ADP-ribose) (PAR). We genetically deleted PARP1 in one of the most widely used human cell systems, i.e. HeLa cells, via TALEN-mediated gene targeting. After comprehensive characterization of these cells during genotoxic stress, we analyzed structure-function relationships of PARP1 by reconstituting PARP1 KO cells with a series of PARP1 variants. Firstly, we verified that the PARP1\E988K mutant exhibits mono-ADP-ribosylation activity and we demonstrate that the PARP1\L713F mutant is constitutively active in cells. Secondly, both mutants exhibit distinct recruitment kinetics to sites of laser-induced DNA damage, which can potentially be attributed to non-covalent PARP1-PAR interaction via several PAR binding motifs. Thirdly, both mutants had distinct functional consequences in cellular patho-physiology, i.e. PARP1\L713F expression triggered apoptosis, whereas PARP1\E988K reconstitution caused a DNA-damage-induced G2 arrest. Importantly, both effects could be rescued by PARP inhibitor treatment, indicating distinct cellular consequences of constitutive PARylation and mono(ADP-ribosyl)ation. Finally, we demonstrate that the cancer-associated PARP1 SNP variant (V762A) as well as a newly identified inherited PARP1 mutation (F304L\V762A) present in a patient with pediatric colorectal carcinoma exhibit altered biochemical and cellular properties, thereby potentially supporting human carcinogenesis. Together, we establish a novel cellular model for PARylation research, by revealing strong structure-function relationships of natural and artificial PARP1 variants.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.