396 results on '"Mullighan, CG"'
Search Results
2. Analytical demands to use whole-genome sequencing in precision oncology
- Author
-
Meggendorfer, M, Jobanputra, V, Wrzeszczynski, KO, Roepman, P, de Bruijn, E, Cuppen, E, Buttner, R, Caldas, C, Grimmond, S, Mullighan, CG, Elemento, O, Rosenquist, R, Schuh, A, Haferlach, T, Meggendorfer, M, Jobanputra, V, Wrzeszczynski, KO, Roepman, P, de Bruijn, E, Cuppen, E, Buttner, R, Caldas, C, Grimmond, S, Mullighan, CG, Elemento, O, Rosenquist, R, Schuh, A, and Haferlach, T
- Abstract
Interrogating the tumor genome in its entirety by whole-genome sequencing (WGS) offers an unprecedented insight into the biology and pathogenesis of cancer, with potential impact on diagnostics, prognostication and therapy selection. WGS is able to detect sequence as well as structural variants and thereby combines central domains of cytogenetics and molecular genetics. Given the potential of WGS in directing targeted therapeutics and clinical decision-making, we envision a gradual transition of the method from research to clinical routine. This review is one out of three within this issue aimed at facilitating this effort, by discussing in-depth analytical validation, clinical interpretation and clinical utility of WGS. The review highlights the requirements for implementing, validating and maintaining a clinical WGS pipeline to obtain high-quality patient-specific data in accordance with the local regulatory landscape. Every step of the WGS pipeline, which includes DNA extraction, library preparation, sequencing, bioinformatics analysis, and data storage, is considered with respect to its logistics, necessities, potential pitfalls, and the required quality management. WGS is likely to drive clinical diagnostics and patient care forward, if requirements and challenges of the technique are recognized and met.
- Published
- 2022
3. Implementation of Whole-Genome and Transcriptome Sequencing Into Clinical Cancer Care
- Author
-
Cuppen, E, Elemento, O, Rosenquist, R, Nikic, S, IJzerman, M, Zaleski, ID, Frederix, G, Levin, L-A, Mullighan, CG, Buettner, R, Pugh, TJ, Grimmond, S, Caldas, C, Andre, F, Custers, I, Campo, E, van Snellenberg, H, Schuh, A, Nakagawa, H, von Kalle, C, Haferlach, T, Froehling, S, Jobanputra, V, Cuppen, E, Elemento, O, Rosenquist, R, Nikic, S, IJzerman, M, Zaleski, ID, Frederix, G, Levin, L-A, Mullighan, CG, Buettner, R, Pugh, TJ, Grimmond, S, Caldas, C, Andre, F, Custers, I, Campo, E, van Snellenberg, H, Schuh, A, Nakagawa, H, von Kalle, C, Haferlach, T, Froehling, S, and Jobanputra, V
- Abstract
PURPOSE: The combination of whole-genome and transcriptome sequencing (WGTS) is expected to transform diagnosis and treatment for patients with cancer. WGTS is a comprehensive precision diagnostic test that is starting to replace the standard of care for oncology molecular testing in health care systems around the world; however, the implementation and widescale adoption of this best-in-class testing is lacking. METHODS: Here, we address the barriers in integrating WGTS for cancer diagnostics and treatment selection and answer questions regarding utility in different cancer types, cost-effectiveness and affordability, and other practical considerations for WGTS implementation. RESULTS: We review the current studies implementing WGTS in health care systems and provide a synopsis of the clinical evidence and insights into practical considerations for WGTS implementation. We reflect on regulatory, costs, reimbursement, and incidental findings aspects of this test. CONCLUSION: WGTS is an appropriate comprehensive clinical test for many tumor types and can replace multiple, cascade testing approaches currently performed. Decreasing sequencing cost, increasing number of clinically relevant aberrations and discovery of more complex biomarkers of treatment response, should pave the way for health care systems and laboratories in implementing WGTS into clinical practice, to transform diagnosis and treatment for patients with cancer.
- Published
- 2022
4. Enhancer retargeting of CDX2 and UBTF::ATXN7L3 define a subtype of high-risk B-progenitor acute lymphoblastic leukemia
- Author
-
Kimura, S, Montefiori, L, Iacobucci, I, Zhao, Y, Gao, Q, Paietta, EM, Haferlach, C, Laird, AD, Mead, PE, Gu, Z, Stock, W, Litzow, M, Rowe, JM, Luger, SM, Hunger, SP, Ryland, GL, Schmidt, B, Ekert, PG, Oshlack, A, Grimmond, SM, Rehn, J, Breen, J, Yeung, D, White, DL, Aldoss, I, Jabbour, EJ, Pui, C-H, Meggendorfer, M, Walter, W, Kern, W, Haferlach, T, Brady, S, Zhang, J, Roberts, KG, Blombery, P, Mullighan, CG, Kimura, S, Montefiori, L, Iacobucci, I, Zhao, Y, Gao, Q, Paietta, EM, Haferlach, C, Laird, AD, Mead, PE, Gu, Z, Stock, W, Litzow, M, Rowe, JM, Luger, SM, Hunger, SP, Ryland, GL, Schmidt, B, Ekert, PG, Oshlack, A, Grimmond, SM, Rehn, J, Breen, J, Yeung, D, White, DL, Aldoss, I, Jabbour, EJ, Pui, C-H, Meggendorfer, M, Walter, W, Kern, W, Haferlach, T, Brady, S, Zhang, J, Roberts, KG, Blombery, P, and Mullighan, CG
- Abstract
Transcriptome sequencing has identified multiple subtypes of B-progenitor acute lymphoblastic leukemia (B-ALL) of prognostic significance, but a minority of cases lack a known genetic driver. Here, we used integrated whole-genome (WGS) and -transcriptome sequencing (RNA-seq), enhancer mapping, and chromatin topology analysis to identify previously unrecognized genomic drivers in B-ALL. Newly diagnosed (n = 3221) and relapsed (n = 177) B-ALL cases with tumor RNA-seq were studied. WGS was performed to detect mutations, structural variants, and copy number alterations. Integrated analysis of histone 3 lysine 27 acetylation and chromatin looping was performed using HiChIP. We identified a subset of 17 newly diagnosed and 5 relapsed B-ALL cases with a distinct gene expression profile and 2 universal and unique genomic alterations resulting from aberrant recombination-activating gene activation: a focal deletion downstream of PAN3 at 13q12.2 resulting in CDX2 deregulation by the PAN3 enhancer and a focal deletion of exons 18-21 of UBTF at 17q21.31 resulting in a chimeric fusion, UBTF::ATXN7L3. A subset of cases also had rearrangement and increased expression of the PAX5 gene, which is otherwise uncommon in B-ALL. Patients were more commonly female and young adult with median age 35 (range,12-70 years). The immunophenotype was characterized by CD10 negativity and immunoglobulin M positivity. Among 16 patients with known clinical response, 9 (56.3%) had high-risk features including relapse (n = 4) or minimal residual disease >1% at the end of remission induction (n = 5). CDX2-deregulated, UBTF::ATXN7L3 rearranged (CDX2/UBTF) B-ALL is a high-risk subtype of leukemia in young adults for which novel therapeutic approaches are required.
- Published
- 2022
5. RUNX2 regulates leukemic cell metabolism and chemotaxis in high-risk T cell acute lymphoblastic leukemia
- Author
-
Matthijssens, F, Sharma, ND, Nysus, M, Nickl, CK, Kang, H, Perez, DR, Lintermans, B, Van Loocke, W, Roels, J, Peirs, S, Demoen, L, Pieters, T, Reunes, L, Lammens, T, De Moerloose, B, Van Nieuwerburgh, F, Deforce, DL, Cheung, LC, Kotecha, RS, Risseeuw, MDP, Van Calenbergh, S, Takarada, T, Yoneda, Y, Van Delft, FW, Lock, RB, Merkley, SD, Chigaev, A, Sklar, LA, Mullighan, CG, Loh, ML, Winter, SS, Hunger, SP, Goossens, S, Castillo, EF, Ornatowski, W, Van Vlierberghe, P, Matlawska-Wasowska, K, Matthijssens, F, Sharma, ND, Nysus, M, Nickl, CK, Kang, H, Perez, DR, Lintermans, B, Van Loocke, W, Roels, J, Peirs, S, Demoen, L, Pieters, T, Reunes, L, Lammens, T, De Moerloose, B, Van Nieuwerburgh, F, Deforce, DL, Cheung, LC, Kotecha, RS, Risseeuw, MDP, Van Calenbergh, S, Takarada, T, Yoneda, Y, Van Delft, FW, Lock, RB, Merkley, SD, Chigaev, A, Sklar, LA, Mullighan, CG, Loh, ML, Winter, SS, Hunger, SP, Goossens, S, Castillo, EF, Ornatowski, W, Van Vlierberghe, P, and Matlawska-Wasowska, K
- Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy with inferior outcome compared with that of B cell ALL. Here, we show that Runt-related transcription factor 2 (RUNX2) was upregulated in high-risk T-ALL with KMT2A rearrangements (KMT2A-R) or an immature immunophenotype. In KMT2A-R cells, we identified RUNX2 as a direct target of the KMT2A chimeras, where it reciprocally bound the KMT2A promoter, establishing a regulatory feed-forward mechanism. Notably, RUNX2 was required for survival of immature and KMT2A-R T-ALL cells in vitro and in vivo. We report direct transcriptional regulation of CXCR4 signaling by RUNX2, thereby promoting chemotaxis, adhesion, and homing to medullary and extramedullary sites. RUNX2 enabled these energy-demanding processes by increasing metabolic activity in T-ALL cells through positive regulation of both glycolysis and oxidative phosphorylation. Concurrently, RUNX2 upregulation increased mitochondrial dynamics and biogenesis in T-ALL cells. Finally, as a proof of concept, we demonstrate that immature and KMT2A-R T-ALL cells were vulnerable to pharmacological targeting of the interaction between RUNX2 and its cofactor CBFβ. In conclusion, we show that RUNX2 acts as a dependency factor in high-risk subtypes of human T-ALL through concomitant regulation of tumor metabolism and leukemic cell migration.
- Published
- 2021
6. Cotargeting BCL-2 and MCL-1 in high-risk B-ALL
- Author
-
Moujalled, DM, Hanna, DT, Hediyeh-zadeh, S, Pomilio, G, Brown, L, Litalien, V, Bartolo, R, Fleming, S, Chanrion, M, Banquet, S, Maragno, A-L, Kraus-Berthier, L, Schoumacher, M, Mullighan, CG, Georgiou, A, White, CA, Lessene, G, Huang, DCS, Roberts, AW, Geneste, O, Rasmussen, L, Davis, MJ, Ekert, PG, Wei, A, Ng, AP, Khaw, SL, Moujalled, DM, Hanna, DT, Hediyeh-zadeh, S, Pomilio, G, Brown, L, Litalien, V, Bartolo, R, Fleming, S, Chanrion, M, Banquet, S, Maragno, A-L, Kraus-Berthier, L, Schoumacher, M, Mullighan, CG, Georgiou, A, White, CA, Lessene, G, Huang, DCS, Roberts, AW, Geneste, O, Rasmussen, L, Davis, MJ, Ekert, PG, Wei, A, Ng, AP, and Khaw, SL
- Abstract
Improving survival outcomes in adult B-cell acute lymphoblastic leukemia (B-ALL) remains a clinical challenge. Relapsed disease has a poor prognosis despite the use of tyrosine kinase inhibitors (TKIs) for Philadelphia chromosome positive (Ph+ ALL) cases and immunotherapeutic approaches, including blinatumomab and chimeric antigen receptor T cells. Targeting aberrant cell survival pathways with selective small molecule BH3-mimetic inhibitors of BCL-2 (venetoclax, S55746), BCL-XL (A1331852), or MCL1 (S63845) is an emerging therapeutic option. We report that combined targeting of BCL-2 and MCL1 is synergistic in B-ALL in vitro. The combination demonstrated greater efficacy than standard chemotherapeutics and TKIs in primary samples from adult B-ALL with Ph+ ALL, Ph-like ALL, and other B-ALL. Moreover, combined BCL-2 or MCL1 inhibition with dasatinib showed potent killing in primary Ph+ B-ALL cases, but the BH3-mimetic combination appeared superior in vitro in a variety of Ph-like ALL samples. In PDX models, combined BCL-2 and MCL1 targeting eradicated ALL from Ph- and Ph+ B-ALL cases, although fatal tumor lysis was observed in some instances of high tumor burden. We conclude that a dual BH3-mimetic approach is highly effective in diverse models of high-risk human B-ALL and warrants assessment in clinical trials that incorporate tumor lysis precautions.
- Published
- 2020
7. Variation in immunoregulatory genes determines the clinical phenotype of common variable immunodeficiency
- Author
-
Mullighan, CG, Marshall, SE, Bunce, M, and Welsh, KI
- Published
- 1999
- Full Text
- View/download PDF
8. OBI-3424, a novel AKR1c3-activated prodrug, exhibits potent efficacy against preclinical models of T-ALL
- Author
-
Evans, K, Duan, JX, Pritchard, T, Jones, CD, McDermott, L, Gu, Z, Toscan, CE, El-Zein, N, Mayoh, C, Erickson, SW, Guo, Y, Meng, F, Jung, D, Rathi, KS, Roberts, KG, Mullighan, CG, Shia, CS, Pearce, T, Teicher, BA, Smith, MA, Lock, RB, Evans, K, Duan, JX, Pritchard, T, Jones, CD, McDermott, L, Gu, Z, Toscan, CE, El-Zein, N, Mayoh, C, Erickson, SW, Guo, Y, Meng, F, Jung, D, Rathi, KS, Roberts, KG, Mullighan, CG, Shia, CS, Pearce, T, Teicher, BA, Smith, MA, and Lock, RB
- Abstract
Purpose: OBI-3424 is a highly selective prodrug that is OBI-3424 significantly prolonged the event-free survival (EFS) converted by aldo-keto reductase family 1 member C3 of nine of nine ALL PDXs by 17.1–77.8 days (treated/control (AKR1C3) to a potent DNA-alkylating agent. OBI-3424 has values 2.5–14.0), and disease regression was observed in eight entered clinical testing for hepatocellular carcinoma and cas-of nine PDXs. A significant reduction (P < 0.0001) in bone trate-resistant prostate cancer, and it represents a potentially marrow infiltration at day 28 was observed in four of six novel treatment for acute lymphoblastic leukemia (ALL). evaluable T-ALL PDXs. The importance of AKR1C3 in the Experimental Design: We assessed AKR1C3 expression by in vivo response to OBI-3424 was verified using a B-ALL PDX RNA-Seq and immunoblotting, and evaluated the in vitro that had been lentivirally transduced to stably overexpress cytotoxicity of OBI-3424. We investigated the pharmacokinet-AKR1C3. OBI-3424 combined with nelarabine resulted in ics of OBI-3424 in mice and nonhuman primates, and assessed prolongation of mouse EFS compared with each single agent the in vivo efficacy of OBI-3424 against a large panel of patient-alone in two T-ALL PDXs. derived xenografts (PDX). Conclusions: OBI-3424 exerted profound in vivo efficacy Results: AKR1C3 mRNA expression was significantly higher against T-ALL PDXs derived predominantly from aggressive in primary T-lineage ALL (T-ALL; n ¼ 264) than B-lineage and fatal disease, and therefore may represent a novel treat-ALL (B-ALL; n ¼ 1,740; P < 0.0001), and OBI-3424 exerted ment for aggressive and chemoresistant T-ALL in an AKR1C3 potent cytotoxicity against T-ALL cell lines and PDXs. In vivo, biomarker-driven clinical trial.
- Published
- 2019
9. Fas gene promoter polymorphisms in primary Sjogren's syndrome
- Author
-
Mullighan, CG, Heatley, S, Lester, S, Rischmueller, M, Gordon, TP, and Bardy, PG
- Subjects
Health - Abstract
Background: Fas mediated apoptosis may be important in the pathogenesis of primary Sjogren's syndrome (pSS). Objective: To examine genetic variation in the promoter region of the Fas gene in pSS. [...]
- Published
- 2004
10. Activating killer-cell immunoglobulin-like receptor haplotype influences clinical outcome following HLA-matched sibling haematopoietic stem cell transplantation
- Author
-
Heatley, SL, Mullighan, CG, Doherty, K, Danner, S, O'Connor, GM, Hahn, U, Szer, J, Schwarer, A, Bradstock, K, Sullivan, LC, Bardy, PG, Brooks, AG, Heatley, SL, Mullighan, CG, Doherty, K, Danner, S, O'Connor, GM, Hahn, U, Szer, J, Schwarer, A, Bradstock, K, Sullivan, LC, Bardy, PG, and Brooks, AG
- Abstract
Natural killer cells are thought to influence the outcome of hematopoietic stem cell transplant (HSCT), impacting on relapse, overall survival, graft versus host disease and the control of infection, in part through the complex interplay between the large and genetically diverse killer immunoglobulin-like receptor (KIR) family and their ligands. This study examined the relationship between KIR gene content and clinical outcomes including the control of opportunistic infections such as cytomegalovirus in the setting of human leucocyte antigen (HLA)-matched sibling HSCT in an Australian cohort. The presence of the KIR B haplotype which contain more activating receptors in the donor, in particular centromeric B haplotype genes (Cen-B), was associated with improved overall survival of patients with acute myeloid leukemia (AML) undergoing sibling HSCT and receiving myeloablative conditioning. Donor Cen-B haplotype was also associated with reduced acute graft versus host disease grades II-IV whereas donor telomeric-B haplotype was associated with decreased incidence of CMV reactivation. In contrast, we were not able to demonstrate a reduced rate of relapse when the donor had KIR Cen-B, however relapse with a donor Cen-A haplotype was a competing risk factor to poor overall survival. Here we show that the presence of donor activating KIR led to improved outcome for the patient, potentially through reduced relapse rates and decreased incidence of acute GvHD translating to improved overall survival. This article is protected by copyright. All rights reserved.
- Published
- 2018
11. The genetic basis and cell of origin of mixed phenotype acute leukaemia.
- Author
-
Alexander, TB, Gu, Z, Iacobucci, I, Dickerson, K, Choi, JK, Xu, B, Payne-Turner, D, Yoshihara, H, Loh, ML, Horan, J, Buldini, B, Basso, G, Elitzur, S, de Haas, V, Zwaan, CM, Yeoh, A, Reinhardt, D, Tomizawa, D, Kiyokawa, N, Lammens, T, De Moerloose, B, Catchpoole, D, Hori, H, Moorman, A, Moore, AS, Hrusak, O, Meshinchi, S, Orgel, E, Devidas, M, Borowitz, M, Wood, B, Heerema, NA, Carrol, A, Yang, Y-L, Smith, MA, Davidsen, TM, Hermida, LC, Gesuwan, P, Marra, MA, Ma, Y, Mungall, AJ, Moore, RA, Jones, SJM, Valentine, M, Janke, LJ, Rubnitz, JE, Pui, C-H, Ding, L, Liu, Y, Zhang, J, Nichols, KE, Downing, JR, Cao, X, Shi, L, Pounds, S, Newman, S, Pei, D, Guidry Auvil, JM, Gerhard, DS, Hunger, SP, Inaba, H, Mullighan, CG, Alexander, TB, Gu, Z, Iacobucci, I, Dickerson, K, Choi, JK, Xu, B, Payne-Turner, D, Yoshihara, H, Loh, ML, Horan, J, Buldini, B, Basso, G, Elitzur, S, de Haas, V, Zwaan, CM, Yeoh, A, Reinhardt, D, Tomizawa, D, Kiyokawa, N, Lammens, T, De Moerloose, B, Catchpoole, D, Hori, H, Moorman, A, Moore, AS, Hrusak, O, Meshinchi, S, Orgel, E, Devidas, M, Borowitz, M, Wood, B, Heerema, NA, Carrol, A, Yang, Y-L, Smith, MA, Davidsen, TM, Hermida, LC, Gesuwan, P, Marra, MA, Ma, Y, Mungall, AJ, Moore, RA, Jones, SJM, Valentine, M, Janke, LJ, Rubnitz, JE, Pui, C-H, Ding, L, Liu, Y, Zhang, J, Nichols, KE, Downing, JR, Cao, X, Shi, L, Pounds, S, Newman, S, Pei, D, Guidry Auvil, JM, Gerhard, DS, Hunger, SP, Inaba, H, and Mullighan, CG
- Abstract
Mixed phenotype acute leukaemia (MPAL) is a high-risk subtype of leukaemia with myeloid and lymphoid features, limited genetic characterization, and a lack of consensus regarding appropriate therapy. Here we show that the two principal subtypes of MPAL, T/myeloid (T/M) and B/myeloid (B/M), are genetically distinct. Rearrangement of ZNF384 is common in B/M MPAL, and biallelic WT1 alterations are common in T/M MPAL, which shares genomic features with early T-cell precursor acute lymphoblastic leukaemia. We show that the intratumoral immunophenotypic heterogeneity characteristic of MPAL is independent of somatic genetic variation, that founding lesions arise in primitive haematopoietic progenitors, and that individual phenotypic subpopulations can reconstitute the immunophenotypic diversity in vivo. These findings indicate that the cell of origin and founding lesions, rather than an accumulation of distinct genomic alterations, prime tumour cells for lineage promiscuity. Moreover, these findings position MPAL in the spectrum of immature leukaemias and provide a genetically informed framework for future clinical trials of potential treatments for MPAL.
- Published
- 2018
12. Conserved IKAROS-regulated genes associated with B-progenitor acute lymphoblastic leukemia outcome
- Author
-
Witkowski, MT, Hu, Y, Roberts, KG, Boer, JM, McKenzie, MD, Liu, GJ, Le Grice, OD, Tremblay, CS, Ghisi, M, Willson, TA, Horstmann, MA, Aifantis, I, Cimmino, L, Frietze, S, den Boer, ML, Mullighan, CG, Smyth, GK, Dickins, RA, Witkowski, MT, Hu, Y, Roberts, KG, Boer, JM, McKenzie, MD, Liu, GJ, Le Grice, OD, Tremblay, CS, Ghisi, M, Willson, TA, Horstmann, MA, Aifantis, I, Cimmino, L, Frietze, S, den Boer, ML, Mullighan, CG, Smyth, GK, and Dickins, RA
- Abstract
Genetic alterations disrupting the transcription factor IKZF1 (encoding IKAROS) are associated with poor outcome in B lineage acute lymphoblastic leukemia (B-ALL) and occur in >70% of the high-risk BCR-ABL1+ (Ph+) and Ph-like disease subtypes. To examine IKAROS function in this context, we have developed novel mouse models allowing reversible RNAi-based control of Ikaros expression in established B-ALL in vivo. Notably, leukemias driven by combined BCR-ABL1 expression and Ikaros suppression rapidly regress when endogenous Ikaros is restored, causing sustained disease remission or ablation. Comparison of transcriptional profiles accompanying dynamic Ikaros perturbation in murine B-ALL in vivo with two independent human B-ALL cohorts identified nine evolutionarily conserved IKAROS-repressed genes. Notably, high expression of six of these genes is associated with inferior event-free survival in both patient cohorts. Among them are EMP1, which was recently implicated in B-ALL proliferation and prednisolone resistance, and the novel target CTNND1, encoding P120-catenin. We demonstrate that elevated Ctnnd1 expression contributes to maintenance of murine B-ALL cells with compromised Ikaros function. These results suggest that IKZF1 alterations in B-ALL leads to induction of multiple genes associated with proliferation and treatment resistance, identifying potential new therapeutic targets for high-risk disease.
- Published
- 2017
13. Genome-Wide Study Links PNPLA3 Variant With Elevated Hepatic Transaminase After Acute Lymphoblastic Leukemia Therapy
- Author
-
Liu, Y, primary, Fernandez, CA, additional, Smith, C, additional, Yang, W, additional, Cheng, C, additional, Panetta, JC, additional, Kornegay, N, additional, Liu, C, additional, Ramsey, LB, additional, Karol, SE, additional, Janke, LJ, additional, Larsen, EC, additional, Winick, N, additional, Carroll, WL, additional, Loh, ML, additional, Raetz, EA, additional, Hunger, SP, additional, Devidas, M, additional, Yang, JJ, additional, Mullighan, CG, additional, Zhang, J, additional, Evans, WE, additional, Jeha, S, additional, Pui, C-H, additional, and Relling, MV, additional
- Published
- 2017
- Full Text
- View/download PDF
14. Synergism of FAK and tyrosine kinase inhibition in Ph+ B-ALL.
- Author
-
Churchman, ML, Evans, K, Richmond, J, Robbins, A, Jones, L, Shapiro, IM, Pachter, JA, Weaver, DT, Houghton, PJ, Smith, MA, Lock, RB, Mullighan, CG, Churchman, ML, Evans, K, Richmond, J, Robbins, A, Jones, L, Shapiro, IM, Pachter, JA, Weaver, DT, Houghton, PJ, Smith, MA, Lock, RB, and Mullighan, CG
- Abstract
BCR-ABL1+ B progenitor acute lymphoblastic leukemia (Ph+ B-ALL) is an aggressive disease that frequently responds poorly to currently available therapies. Alterations in IKZF1, which encodes the lymphoid transcription factor Ikaros, are present in over 80% of Ph+ ALL and are associated with a stem cell-like phenotype, aberrant adhesion molecule expression and signaling, leukemic cell adhesion to the bone marrow stem cell niche, and poor outcome. Here, we show that FAK1 is upregulated in Ph+ B-ALL with further overexpression in IKZF1-altered cells and that the FAK inhibitor VS-4718 potently inhibits aberrant FAK signaling and leukemic cell adhesion, potentiating responsiveness to tyrosine kinase inhibitors, inducing cure in vivo. Thus, targeting FAK with VS-4718 is an attractive approach to overcome the deleterious effects of FAK overexpression in Ph+ B-ALL, particularly in abrogating the adhesive phenotype induced by Ikaros alterations, and warrants evaluation in clinical trials for Ph+ B-ALL, regardless of IKZF1 status.
- Published
- 2016
15. Characterization of leukemias with ETV6-ABL1 fusion
- Author
-
Zaliova, M, Moorman, A, Cazzaniga, G, Stanulla, M, Harvey, R, Roberts, K, Heatley, S, Loh, M, Konopleva, M, Chen, I, Zimmermannova, O, Schwab, C, Smith, O, Mozziconacci, M, Chabannon, C, Kim, M, Frederik Falkenburg, J, Norton, A, Marshall, K, Haas, O, Starkova, J, Stuchly, J, Hunger, S, White, D, Mullighan, C, Willman, C, Stary, J, Trka, J, Zuna, J, Moorman, AV, Harvey, RC, Roberts, KG, Heatley, SL, Loh, ML, Chen, IM, Mozziconacci, MJ, Frederik Falkenburg, JH, Haas, OA, Hunger, SP, Mullighan, CG, Willman, CL, Zaliova, M, Moorman, A, Cazzaniga, G, Stanulla, M, Harvey, R, Roberts, K, Heatley, S, Loh, M, Konopleva, M, Chen, I, Zimmermannova, O, Schwab, C, Smith, O, Mozziconacci, M, Chabannon, C, Kim, M, Frederik Falkenburg, J, Norton, A, Marshall, K, Haas, O, Starkova, J, Stuchly, J, Hunger, S, White, D, Mullighan, C, Willman, C, Stary, J, Trka, J, Zuna, J, Moorman, AV, Harvey, RC, Roberts, KG, Heatley, SL, Loh, ML, Chen, IM, Mozziconacci, MJ, Frederik Falkenburg, JH, Haas, OA, Hunger, SP, Mullighan, CG, and Willman, CL
- Abstract
To characterize the incidence, clinical features and genetics of ETV6-ABL1 leukemias, representing targetable kinase-activating lesions, we analyzed 44 new and published cases of ETV6-ABL1-positive hematologic malignancies [22 cases of acute lymphoblastic leukemia (13 children, 9 adults) and 22 myeloid malignancies (18 myeloproliferative neoplasms, 4 acute myeloid leukemias)]. The presence of the ETV6-ABL1 fusion was ascertained by cytogenetics, fluorescence in-situ hybridization, reverse transcriptase-polymerase chain reaction and RNA sequencing. Genomic and gene expression profiling was performed by single nucleotide polymorphism and expression arrays. Systematic screening of more than 4,500 cases revealed that in acute lymphoblastic leukemia ETV6-ABL1 is rare in childhood (0.17% cases) and slightly more common in adults (0.38%). There is no systematic screening of myeloproliferative neoplasms; however, the number of ETV6-ABL1-positive cases and the relative incidence of acute lymphoblastic leukemia and myeloproliferative neoplasms suggest that in adulthood ETV6-ABL1 is more common in BCR-ABL1-negative chronic myeloid leukemia-like myeloproliferations than in acute lymphoblastic leukemia. The genomic profile of ETV6-ABL1 acute lymphoblastic leukemia resembled that of BCR-ABL1 and BCR-ABL1- like cases with 80% of patients having concurrent CDKN2A/B and IKZF1 deletions. In the gene expression profiling all the ETV6-ABL1-positive samples clustered in close vicinity to BCR-ABL1 cases. All but one of the cases of ETV6- ABL1 acute lymphoblastic leukemia were classified as BCR-ABL1-like by a standardized assay. Over 60% of patients died, irrespectively of the disease or age subgroup examined. In conclusion, ETV6-ABL1 fusion occurs in both lymphoid and myeloid leukemias; the genomic profile and clinical behavior resemble BCR-ABL1-positive malignancies, including the unfavorable prognosis, particularly of acute leukemias. The poor outcome suggests that treatment with tyros
- Published
- 2016
16. Comparison of genome sequencing and clinical genotyping for pharmacogenes
- Author
-
Yang, W, primary, Wu, G, additional, Broeckel, U, additional, Smith, CA, additional, Turner, V, additional, Haidar, CE, additional, Wang, S, additional, Carter, R, additional, Karol, SE, additional, Neale, G, additional, Crews, KR, additional, Yang, JJ, additional, Mullighan, CG, additional, Downing, JR, additional, Evans, WE, additional, and Relling, MV, additional
- Published
- 2016
- Full Text
- View/download PDF
17. Evaluation of the in vitro and in vivo efficacy of the JAK inhibitor AZD1480 against JAK-mutated acute lymphoblastic leukemia
- Author
-
Suryani, S, Bracken, LS, Harvey, RC, Sia, KCS, Carol, H, Chen, IM, Evans, K, Dietrich, PA, Roberts, KG, Kurmasheva, RT, Billups, CA, Mullighan, CG, Willman, CL, Loh, ML, Hunger, SP, Houghton, PJ, Smith, MA, Lock, RB, Suryani, S, Bracken, LS, Harvey, RC, Sia, KCS, Carol, H, Chen, IM, Evans, K, Dietrich, PA, Roberts, KG, Kurmasheva, RT, Billups, CA, Mullighan, CG, Willman, CL, Loh, ML, Hunger, SP, Houghton, PJ, Smith, MA, and Lock, RB
- Abstract
Genome-wide studies have identified a high-risk subgroup of pediatric acute lymphoblastic leukemia (ALL) harboring mutations in the Janus kinases (JAK). The purpose of this study was to assess the preclinical efficacy of the JAK1/2 inhibitor AZD1480, both as a single agent and in combination with the MEK inhibitor selumetinib, against JAK-mutated patient-derived xenografts. Patient-derived xenografts were established in immunodeficient mice from bone marrow or peripheral blood biopsy specimens, and their gene expression profiles compared with the original patient biopsies by microarray analysis. JAK/STAT and MAPK signaling pathways, and the inhibitory effects of targeted drugs, were interrogated by immunoblotting of phosphoproteins. The antileukemic effects of AZD1480 and selumetinib, alone and in combination, were tested against JAK-mutated ALL xenografts both in vitro and in vivo. Xenografts accurately represented the primary disease as determined by gene expression profiling. Cellular phosphoprotein analysis demonstrated that JAK-mutated xenografts exhibited heightened activation status of JAK/STAT and MAPK signaling pathways compared with typical B-cell precursor ALL xenografts, which were inhibited by AZD1480 exposure. However, AZD1480 exhibited modest single-agent in vivo efficacy against JAK-mutated xenografts. Combining AZD1480 with selumetinib resulted in profound synergistic in vitro cell killing, although these results were not translated in vivo despite evidence of target inhibition. Despite validation of target inhibition and the demonstration of profound in vitro synergy between AZD1480 and selumetinib, it is likely that prolonged target inhibition is required to achieve in vivo therapeutic enhancement between JAK and MEK inhibitors in the treatment of JAK-mutated ALL.
- Published
- 2015
18. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias.
- Author
-
Andersson, AK, Ma, J, Wang, J, Chen, X, Gedman, AL, Dang, J, Nakitandwe, J, Holmfeldt, L, Parker, M, Easton, J, Huether, R, Kriwacki, R, Rusch, M, Wu, G, Li, Y, Mulder, H, Raimondi, S, Pounds, S, Kang, G, Shi, L, Becksfort, J, Gupta, P, Payne-Turner, D, Vadodaria, B, Boggs, K, Yergeau, D, Manne, J, Song, G, Edmonson, M, Nagahawatte, P, Wei, L, Cheng, C, Pei, D, Sutton, R, Venn, NC, Chetcuti, A, Rush, A, Catchpoole, D, Heldrup, J, Fioretos, T, Lu, C, Ding, L, Pui, C-H, Shurtleff, S, Mullighan, CG, Mardis, ER, Wilson, RK, Gruber, TA, Zhang, J, Downing, JR, St. Jude Children's Research Hospital–Washington University Pediatric Cancer Genome Project, Andersson, AK, Ma, J, Wang, J, Chen, X, Gedman, AL, Dang, J, Nakitandwe, J, Holmfeldt, L, Parker, M, Easton, J, Huether, R, Kriwacki, R, Rusch, M, Wu, G, Li, Y, Mulder, H, Raimondi, S, Pounds, S, Kang, G, Shi, L, Becksfort, J, Gupta, P, Payne-Turner, D, Vadodaria, B, Boggs, K, Yergeau, D, Manne, J, Song, G, Edmonson, M, Nagahawatte, P, Wei, L, Cheng, C, Pei, D, Sutton, R, Venn, NC, Chetcuti, A, Rush, A, Catchpoole, D, Heldrup, J, Fioretos, T, Lu, C, Ding, L, Pui, C-H, Shurtleff, S, Mullighan, CG, Mardis, ER, Wilson, RK, Gruber, TA, Zhang, J, Downing, JR, and St. Jude Children's Research Hospital–Washington University Pediatric Cancer Genome Project
- Abstract
Infant acute lymphoblastic leukemia (ALL) with MLL rearrangements (MLL-R) represents a distinct leukemia with a poor prognosis. To define its mutational landscape, we performed whole-genome, exome, RNA and targeted DNA sequencing on 65 infants (47 MLL-R and 18 non-MLL-R cases) and 20 older children (MLL-R cases) with leukemia. Our data show that infant MLL-R ALL has one of the lowest frequencies of somatic mutations of any sequenced cancer, with the predominant leukemic clone carrying a mean of 1.3 non-silent mutations. Despite this paucity of mutations, we detected activating mutations in kinase-PI3K-RAS signaling pathway components in 47% of cases. Surprisingly, these mutations were often subclonal and were frequently lost at relapse. In contrast to infant cases, MLL-R leukemia in older children had more somatic mutations (mean of 6.5 mutations/case versus 1.3 mutations/case, P = 7.15 × 10(-5)) and had frequent mutations (45%) in epigenetic regulators, a category of genes that, with the exception of MLL, was rarely mutated in infant MLL-R ALL.
- Published
- 2015
19. Acute lymphoblastic leukemia in children with Down syndrome: A retrospective analysis from the Ponte di Legno study group
- Author
-
Buitenkamp, T, Izraeli, S, Zimmermann, M, Forestier, E, Heerema, N, van den Heuvel Eibrink, M, Pieters, R, Korbijn, C, Silverman, L, Schmiegelow, K, Liang, D, Horibe, K, Arico, M, Biondi, A, Basso, G, Rabin, K, Schrappe, M, Cario, G, Mann, G, Morak, M, Panzer Grümayer, R, Mondelaers, V, Lammens, T, Cavé, H, Stark, B, Ganmore, I, Moorman, A, Vora, A, Hunger, S, Pui, C, Mullighan, C, Manabe, A, Escherich, G, Kowalczyk, J, Whitlock, J, Zwaan, C, Buitenkamp, TD, Heerema, NA, van den Heuvel Eibrink, MM, Korbijn, CM, Silverman, LB, Liang, DC, Rabin, KR, Moorman, AV, Hunger, SP, Pui, CH, Mullighan, CG, Kowalczyk, JR, Whitlock, JA, Zwaan, CM, BIONDI, ANDREA, Buitenkamp, T, Izraeli, S, Zimmermann, M, Forestier, E, Heerema, N, van den Heuvel Eibrink, M, Pieters, R, Korbijn, C, Silverman, L, Schmiegelow, K, Liang, D, Horibe, K, Arico, M, Biondi, A, Basso, G, Rabin, K, Schrappe, M, Cario, G, Mann, G, Morak, M, Panzer Grümayer, R, Mondelaers, V, Lammens, T, Cavé, H, Stark, B, Ganmore, I, Moorman, A, Vora, A, Hunger, S, Pui, C, Mullighan, C, Manabe, A, Escherich, G, Kowalczyk, J, Whitlock, J, Zwaan, C, Buitenkamp, TD, Heerema, NA, van den Heuvel Eibrink, MM, Korbijn, CM, Silverman, LB, Liang, DC, Rabin, KR, Moorman, AV, Hunger, SP, Pui, CH, Mullighan, CG, Kowalczyk, JR, Whitlock, JA, Zwaan, CM, and BIONDI, ANDREA
- Abstract
Children with Down syndrome (DS) have an increased risk of B-cell precursor (BCP) acute lymphoblastic leukemia (ALL). The prognostic factors and outcome of DS-ALL patients treated in contemporary protocols are uncertain. We studied 653 DS-ALL patients enrolled in 16 international trials from 1995 to 2004. Non-DS BCP-ALL patients from the Dutch Child Oncology Group and Berlin-Frankfurt- Münster were reference cohorts. DS-ALL patients had a higher 8-year cumulative incidence of relapse (26% ± 2% vs 15% ± 1%, P < .001) and 2-year treatment-related mortality (TRM) (7% ± 1% vs 2.0% ± <1%, P < .0001) than non-DS patients, resulting in lower 8-year event-free survival (EFS) (64% ± 2% vs 81% ± 2%, P < .0001) and overall survival (74% ± 2% vs 89% ± 1%, P < .0001). Independent favorable prognostic factors include age <6 years (hazard ratio [HR] = 0.58, P = .002), white blood cell (WBC) count <10 3 109/L (HR = 0.60, P = .005), and ETV6-RUNX1 (HR = 0.14, P = .006) for EFS and age (HR = 0.48, P < .001), ETV6-RUNX1 (HR = 0.1, P = .016) and high hyperdiploidy (HeH) (HR = 0.29, P = .04) for relapse-free survival. TRM was the major cause of death in ETV6-RUNX1 and HeH DSALLs. Thus, while relapse is the main contributor to poorer survival in DS-ALL, infection-associated TRM was increased in all protocol elements, unrelated to treatment phase or regimen. Future strategies to improve outcome in DS-ALL should include improved supportive care throughout therapy and reduction of therapy in newly identified good-prognosis subgroups. (Blood. 2014; 123(1):70-77). © 2014 by The American Society of Hematology.
- Published
- 2014
20. A recurrent germline PAX5 mutation confers susceptibility to pre-B cell acute lymphoblastic leukemia
- Author
-
Shah, S, Schrader, KA, Vijai, J, Miething, C, Hayes, J, Klein, RJ, Gao, X, Kitzing, T, Lowe, SW, Offit, K, Littman, J, Offit, L, Rau-Murthy, R, Fleischut, MH, Corines, M, Manschreck, C, Waanders, E, Wei, L, Ma, J, Chen, S-C, Song, G, Cheng, J, Raimondi, SC, Roberts, KG, Downing, JR, Mullighan, CG, Kuiper, RP, Simons, A, Timms, AE, Wechsler, J, Horwitz, MS, Yang, J, Zhang, J, Wu, G, Rusch, M, Nagahawatte, P, Meyers, P, Bhojwani, D, Sandlund, JT, Jhanwar, S, Murali, R, Maslak, P, Fleisher, M, Murty, VV, Schiffman, JD, Onel, K, Plon, SE, Wheeler, DA, Ritter, D, Ziegler, DS, Sutton, R, Tucker, K, Chenevix-Trench, G, Li, J, Huntsman, DG, Hansford, S, Senz, J, Walsh, T, Lee, M, King, M-C, Hahn, CN, Scott, HS, Lo, SM, Levine, RL, Viale, A, Socci, ND, Nathanson, KL, Daly, M, Lipkin, SM, Altshuler, D, Shah, S, Schrader, KA, Vijai, J, Miething, C, Hayes, J, Klein, RJ, Gao, X, Kitzing, T, Lowe, SW, Offit, K, Littman, J, Offit, L, Rau-Murthy, R, Fleischut, MH, Corines, M, Manschreck, C, Waanders, E, Wei, L, Ma, J, Chen, S-C, Song, G, Cheng, J, Raimondi, SC, Roberts, KG, Downing, JR, Mullighan, CG, Kuiper, RP, Simons, A, Timms, AE, Wechsler, J, Horwitz, MS, Yang, J, Zhang, J, Wu, G, Rusch, M, Nagahawatte, P, Meyers, P, Bhojwani, D, Sandlund, JT, Jhanwar, S, Murali, R, Maslak, P, Fleisher, M, Murty, VV, Schiffman, JD, Onel, K, Plon, SE, Wheeler, DA, Ritter, D, Ziegler, DS, Sutton, R, Tucker, K, Chenevix-Trench, G, Li, J, Huntsman, DG, Hansford, S, Senz, J, Walsh, T, Lee, M, King, M-C, Hahn, CN, Scott, HS, Lo, SM, Levine, RL, Viale, A, Socci, ND, Nathanson, KL, Daly, M, Lipkin, SM, and Altshuler, D
- Abstract
Somatic alterations of the lymphoid transcription factor gene PAX5 (also known as BSAP) are a hallmark of B cell precursor acute lymphoblastic leukemia (B-ALL), but inherited mutations of PAX5 have not previously been described. Here we report a new heterozygous germline variant, c.547G>A (p.Gly183Ser), affecting the octapeptide domain of PAX5 that was found to segregate with disease in two unrelated kindreds with autosomal dominant B-ALL. Leukemic cells from all affected individuals in both families exhibited 9p deletion, with loss of heterozygosity and retention of the mutant PAX5 allele at 9p13. Two additional sporadic ALL cases with 9p loss harbored somatic PAX5 substitutions affecting Gly183. Functional and gene expression analysis of the PAX5 mutation demonstrated that it had significantly reduced transcriptional activity. These data extend the role of PAX5 alterations in the pathogenesis of pre-B cell ALL and implicate PAX5 in a new syndrome of susceptibility to pre-B cell neoplasia. © 2013 Nature America, Inc. All rights reserved.
- Published
- 2013
21. Combined Targeting of JAK2 and Bcl-2/Bcl-xL to Cure Mutant JAK2-Driven Malignancies and Overcome Acquired Resistance to JAK2 Inhibitors
- Author
-
Waibel, M, Solomon, VS, Knight, DA, Ralli, RA, Kim, SK, Banks, KM, Vidacs, E, Virely, C, Sia, KCS, Bracken, LS, Collins-Underwood, R, Drenberg, C, Ramsey, LB, Meyer, SC, Takiguchi, M, Dickins, RA, Levine, R, Ghysdael, J, Dawson, MA, Lock, RB, Mullighan, CG, Johnstone, RW, Waibel, M, Solomon, VS, Knight, DA, Ralli, RA, Kim, SK, Banks, KM, Vidacs, E, Virely, C, Sia, KCS, Bracken, LS, Collins-Underwood, R, Drenberg, C, Ramsey, LB, Meyer, SC, Takiguchi, M, Dickins, RA, Levine, R, Ghysdael, J, Dawson, MA, Lock, RB, Mullighan, CG, and Johnstone, RW
- Abstract
To design rational therapies for JAK2-driven hematological malignancies, we functionally dissected the key survival pathways downstream of hyperactive JAK2. In tumors driven by mutant JAK2, Stat1, Stat3, Stat5, and the Pi3k and Mek/Erk pathwayswere constitutively active, and gene expressionprofiling of TEL-JAK2 T-ALL cells revealed theupregulation of prosurvival Bcl-2 family genes. Combining the Bcl-2/Bcl-xL inhibitor ABT-737 with JAK2 inhibitors mediated prolonged disease regressions and cures in mice bearing primary human andmouse JAK2 mutant tumors. Moreover, combined targeting of JAK2 and Bcl-2/Bcl-xL was ableto circumvent and overcome acquired resistance to single-agent JAK2 inhibitor treatment. Thus, inhibiting the oncogenic JAK2 signaling network at two nodal points, at the initiating stage (JAK2) and the effector stage (Bcl-2/Bcl-xL), is highly effectiveand provides a clearly superior therapeutic benefitthan targeting just one node. Therefore, we have defined a potentially curative treatment for hematological malignancies expressing constitutively active JAK2.
- Published
- 2013
22. Highly sensitive MRD tests for ALL based on the IKZF1 Delta 3-6 microdeletion
- Author
-
Venn, NC, Van der velden, VHJ, De bie, M, Waanders, E, Giles, JE, Law, T, Kuiper, RP, de Haas, V, Mullighan, CG, Haber, M, Marshall, GM, Norris, MD, Van dongen, JJM, Sutton, R, Giles, J, Venn, NC, Van der velden, VHJ, De bie, M, Waanders, E, Giles, JE, Law, T, Kuiper, RP, de Haas, V, Mullighan, CG, Haber, M, Marshall, GM, Norris, MD, Van dongen, JJM, Sutton, R, and Giles, J
- Abstract
No abstract available.
- Published
- 2012
23. Analysis of the chronic lymphocytic leukemia coding genome: role of NOTCH1 mutational activation
- Author
-
Fabbri, G, Rasi, S, Rossi, D, Trifonov, V, Khiabanian, H, Ma, J, Grunn, A, Fangazio, M, Capello, D, Monti, S, Cresta, S, Gargiulo, E, Forconi, F, Guarini, A, Arcaini, L, Paulli, M, Laurenti, Luca, Larocca, Luigi Maria, Marasca, R, Gattei, V, Oscier, D, Bertoni, Francesco, Mullighan, Cg, Foá, R, Pasqualucci, L, Rabadan, R, Dalla-Favera, R, Gaidano, G, Monti, Stefano, Laurenti, Luca (ORCID:0000-0002-8327-1396), Larocca, Luigi Maria (ORCID:0000-0003-1739-4758), Fabbri, G, Rasi, S, Rossi, D, Trifonov, V, Khiabanian, H, Ma, J, Grunn, A, Fangazio, M, Capello, D, Monti, S, Cresta, S, Gargiulo, E, Forconi, F, Guarini, A, Arcaini, L, Paulli, M, Laurenti, Luca, Larocca, Luigi Maria, Marasca, R, Gattei, V, Oscier, D, Bertoni, Francesco, Mullighan, Cg, Foá, R, Pasqualucci, L, Rabadan, R, Dalla-Favera, R, Gaidano, G, Monti, Stefano, Laurenti, Luca (ORCID:0000-0002-8327-1396), and Larocca, Luigi Maria (ORCID:0000-0003-1739-4758)
- Abstract
The pathogenesis of chronic lymphocytic leukemia (CLL), the most common leukemia in adults, is still largely unknown. The full spectrum of genetic lesions that are present in the CLL genome, and therefore the number and identity of dysregulated cellular pathways, have not been identified. By combining next-generation sequencing and copy number analysis, we show here that the typical CLL coding genome contains <20 clonally represented gene alterations/case, including predominantly nonsilent mutations, and fewer copy number aberrations. These analyses led to the discovery of several genes not previously known to be altered in CLL. Although most of these genes were affected at low frequency in an expanded CLL screening cohort, mutational activation of NOTCH1, observed in 8.3% of CLL at diagnosis, was detected at significantly higher frequency during disease progression toward Richter transformation (31.0%), as well as in chemorefractory CLL (20.8%). Consistent with the association of NOTCH1 mutations with clinically aggressive forms of the disease, NOTCH1 activation at CLL diagnosis emerged as an independent predictor of poor survival. These results provide initial data on the complexity of the CLL coding genome and identify a dysregulated pathway of diagnostic and therapeutic relevance.
- Published
- 2011
24. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia
- Author
-
Coustan Smith, E, Mullighan, C, Onciu, M, Behm, F, Raimondi, S, Pei, D, Cheng, C, Su, X, Rubnitz, J, Basso, G, Biondi, A, Pui, C, Downing, J, Campana, D, Mullighan, CG, Behm, FG, Raimondi, SC, Rubnitz, JE, Pui, CH, Downing, JR, Campana, D., BIONDI, ANDREA, Coustan Smith, E, Mullighan, C, Onciu, M, Behm, F, Raimondi, S, Pei, D, Cheng, C, Su, X, Rubnitz, J, Basso, G, Biondi, A, Pui, C, Downing, J, Campana, D, Mullighan, CG, Behm, FG, Raimondi, SC, Rubnitz, JE, Pui, CH, Downing, JR, Campana, D., and BIONDI, ANDREA
- Abstract
Background: About a fifth of children with acute T-lymphoblastic leukaemia (T-ALL) succumb to the disease, suggesting an unrecognised biological heterogeneity that might contribute to drug resistance. We postulated that T-ALL originating from early T-cell precursors (ETPs), a recently defined subset of thymocytes that retain stem-cell-like features, would respond poorly to lymphoid-cell-directed therapy. We studied leukaemic cells, collected at diagnosis, to identify cases with ETP features and determine their clinical outcome. Methods: Leukaemic cells from 239 patients with T-ALL enrolled at St Jude Children's Research Hospital (n=139) and in the Italian national study Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP) ALL-2000 (n=100) were assessed by gene-expression profiling, flow cytometry, and single nucleotide polymorphism array analysis. Probabilities of survival and treatment failure were calculated for subgroups considered to have ETP-ALL or typical T-ALL. Findings: 30 patients (12·6%) had leukaemic lymphoblasts with an ETP-related gene-expression signature or its associated distinctive immunophenotype (CD1a-, CD8-, CD5weak with stem-cell or myeloid markers). Cases of ETP-ALL showed increased genomic instability, in terms of number and size of gene lesions, compared with those with typical T-ALL. Patients with this form of leukaemia had high risk of remission failure or haematological relapse (72% [95% CI 40-100] at 10 years vs 10% [4-16] at 10 years for patients with typical T-ALL treated at St Jude Children's Research Hospital; and 57% [25-89] at 2 years vs 14% [6-22] at 2 years for patients treated in the AIEOP trial). Interpretation: ETP-ALL is a distinct, previously unrecognised, pathobiological entity that confers a poor prognosis with use of standard intensive chemotherapy. Its early recognition, by use of the gene expression and immunophenotypic criteria outlined here, is essential for the development of an effective clinical management
- Published
- 2009
25. HEPATITIS C AFTER INTRAVENOUS IMMUNOGLOBULIN
- Author
-
Sewell, WAC, primary, Mullighan, CG, additional, Christie, J, additional, and Chapel, HM, additional
- Published
- 1997
- Full Text
- View/download PDF
26. Absence of biallelic TCRgamma deletion predicts early treatment failure in pediatric T-cell acute lymphoblastic leukemia.
- Author
-
Gutierrez A, Dahlberg SE, Neuberg DS, Zhang J, Grebliunaite R, Sanda T, Protopopov A, Tosello V, Kutok J, Larson RS, Borowitz MJ, Loh ML, Ferrando AA, Winter SS, Mullighan CG, Silverman LB, Chin L, Hunger SP, Sallan SE, and Look AT
- Published
- 2010
- Full Text
- View/download PDF
27. Mannose-binding lectin deficiency does not increase the prevalence of Helicobacter pylori seropositivity.
- Author
-
Worthley DL, Mullighan CG, Dean MM, Gordon DL, Phillips P, Heatley S, Young GP, Bardy PG, Worthley, Daniel L, Mullighan, Charles G, Dean, Melinda M, Gordon, David L, Phillips, Peter, Heatley, Susan, Young, Graeme P, and Bardy, Peter G
- Published
- 2007
- Full Text
- View/download PDF
28. Highly sensitive MRD tests for ALL based on the IKZF1 Δ3-6 microdeletion.
- Author
-
Venn NC, van der Velden VH, de Bie M, Waanders E, Giles JE, Law T, Kuiper RP, de Haas V, Mullighan CG, Haber M, Marshall GM, Md N, van Dongen JJ, Sutton R, Venn, N C, van der Velden, V H J, de Bie, M, Waanders, E, Giles, J E, and Law, T
- Published
- 2012
- Full Text
- View/download PDF
29. JAK2 -- a new player in acute lymphoblastic leukaemia.
- Author
-
Mullighan CG
- Published
- 2008
- Full Text
- View/download PDF
30. Rearrangement of CRLF2 in B-progenitor- and Down syndrome-associated acute lymphoblastic leukemia
- Author
-
Richard T. Williams, Julia Meyer, Nyla A. Heerema, Charles G. Mullighan, Stephen P. Hunger, Jing Ma, William L. Carroll, Cheryl L. Willman, Fady M. Mikhail, Wei Liu, Giuseppe Basso, Letha A. Phillips, Jinjun Cheng, Andrew J. Carroll, Jinghui Zhang, Richard C. Harvey, Andrea Pession, Karen R. Rabin, Susana C. Raimondi, Elaine Coustan-Smith, Michael G. Loudin, J. Racquel Collins-Underwood, James R. Downing, Ching-Hon Pui, Mullighan CG, Collins-Underwood JR, Phillips LA, Loudin MG, Liu W, Zhang J, Ma J, Coustan-Smith E, Harvey RC, Willman CL, Mikhail FM, Meyer J, Carroll AJ, Williams RT, Cheng J, Heerema NA, Basso G, Pession A, Pui CH, Raimondi SC, Hunger SP, Downing JR, Carroll WL, and Rabin KR.
- Subjects
Down syndrome ,Recombinant Fusion Proteins ,Pseudoautosomal region ,Aneuploidy ,Chromosomal translocation ,Biology ,Article ,Cell Line ,Exon ,Mice ,Acute lymphocytic leukemia ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Genetics ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Receptors, Cytokine ,Gene Rearrangement ,Acute leukemia ,Base Sequence ,Receptors, Purinergic P2 ,Gene rearrangement ,Janus Kinase 2 ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Cancer research ,Chromosome Deletion ,Down Syndrome - Abstract
Aneuploidy and translocations are hallmarks of B-progenitor acute lymphoblastic leukemia (ALL), but many patients lack a recurring chromosomal alteration. Here we report a recurring interstitial deletion of the pseudoautosomal region 1 of chromosomes X and Y in B-progenitor ALL that juxtaposes the first, non-coding exon of P2RY8 to the coding region of CRLF2 (which encodes cytokine receptor like factor 2, or thymic stromal lymphopoietin receptor). The P2RY8-CRLF2 fusion was identified in 7% of B-progenitor ALL cases, and was identified in over 50% of ALL cases arising in patients with Down syndrome (53% of 75 cases). CRLF2 alteration was associated with the presence of activating JAK mutations, and expression of P2RY8-CRLF2 together with JAK2 mutants resulted in constitutive Jak-Stat activation and cytokine-independent growth of Ba/F3-IL7R cells, indicating that these two genetic lesions together contribute to leukemogenesis in B-progenitor ALL.
- Published
- 2009
31. Genomic profiling of circulating tumor DNA for childhood cancers.
- Author
-
Lei S, Jia S, Takalkar S, Chang TC, Ma X, Szlachta K, Xu K, Cheng Z, Hui Y, Koo SC, Mead PE, Gao Q, Kumar P, Bailey CP, Sunny J, Pappo AS, Federico SM, Robinson GW, Gajjar A, Rubnitz JE, Jeha S, Pui CH, Inaba H, Wu G, Klco JM, Tatevossian RG, and Mullighan CG
- Abstract
The utility of circulating tumor DNA (ctDNA) analysis has not been well-established for disease detection and monitoring of childhood cancers, especially leukemias. We developed PeCan-Seq, a deep sequencing method targeting diverse somatic genomic variants in cell-free samples in childhood cancer. Plasma samples were collected at diagnosis from 233 children with hematologic, solid and brain tumors. All children with hematologic malignancy (n = 177) had detectable ctDNA at diagnosis. The median ctDNA fraction was 0.77, and 97% of 789 expected tumor variants were identified, including sequence mutations, copy number variations, and structural variations responsible for oncogenic fusions. In contrast, ctDNA was detected in 19 of 38 solid tumor patients and 1 of 18 brain tumor patients. Somatic variants from ctDNA were correlated with minimal residual disease levels as determined by flow cytometry in serial plasma samples from patients with B-cell acute lymphoblastic leukemia (B-ALL). We showcase multi-tumor detection by ctDNA analysis for a patient with concurrent B-ALL and neuroblastoma. In conclusion, PeCan-seq sensitively identified heterogeneous ctDNA alterations from 1 mL plasma for childhood hematologic malignancies and a subset of solid tumors. PeCan-seq provides a robust, non-invasive approach to augment comprehensive genomic profiling at diagnosis and mutation-specific detection during disease monitoring., Competing Interests: Competing interests This work was supported by funding from the American Lebanese Syrian Associated Charities to St. Jude Children’s Research Hospital, a National Cancer Institute R35 grant (CA197695) to CGM, and a National Cancer Institute P30 grant (CA021765) to the Hartwell Center., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
32. Single-cell transcriptional mapping reveals genetic and non-genetic determinants of aberrant differentiation in AML.
- Author
-
Zeng AGX, Iacobucci I, Shah S, Mitchell A, Wong G, Bansal S, Chen D, Gao Q, Kim H, Kennedy JA, Arruda A, Minden MD, Haferlach T, Mullighan CG, and Dick JE
- Abstract
In acute myeloid leukemia (AML), genetic mutations distort hematopoietic differentiation, resulting in the accumulation of leukemic blasts. Yet, it remains unclear how these mutations intersect with cellular origins and whether they converge upon similar differentiation patterns. Single-cell RNA sequencing (scRNA-seq) has enabled high-resolution mapping of the relationship between leukemia and normal cell states, yet this application is hampered by imprecise reference maps of normal hematopoiesis and small sample sizes among patient cohorts. As a first step we constructed a reference atlas of human bone marrow hematopoiesis from 263,519 single-cell transcriptomes spanning 55 cellular states, that was benchmarked against independent datasets of immunophenotypically pure hematopoietic stem and progenitor cells. Using this reference atlas, we mapped over 1.2 million single-cell transcriptomes spanning 318 AML, mixed phenotype acute leukemia (MPAL), and acute erythroid leukemia (AEL) samples. This large-scale analysis, together with systematic mapping of genotype-to-phenotype associations between driver mutations and differentiation landscapes, revealed convergence of diverse genetic alterations on twelve recurrent patterns of aberrant differentiation in AML. This included unconventional lymphoid and erythroid priming linked to RUNX1 and TP53 mutations, respectively. We also identified non-genetic determinants of AML differentiation such as two subgroups of KMT2A -rearranged AML that differ in the identity of their leukemic stem cells (LSCs), likely reflecting distinct cellular origins. Furthermore, distinct LSC-driven hierarchies can co-exist within individual patients, providing insights into AML evolution. Together, precise mapping of normal and malignant cell states provides a framework for advancing the study and disease classification of hematologic malignancies thereby informing therapy development., Competing Interests: Disclosure of Conflicts of Interest J.E.D.: received research funding from BMS/Celgene and IP licenses from Pfizer/Trillium Therapeutics. I.I.: reported consultation honoraria from Arima, travel expenses reimbursed by Mission Bio for invited talk and honoraria from MD Education. C.G.M.: received research funding from AbbVie and Pfizer, honoraria from Amgen and Illumina, royalty payments from Cyrus, and is on an advisory board for Illumina. T.H.: equity ownership of MLL Munich Leukemia Laboratory.
- Published
- 2024
- Full Text
- View/download PDF
33. MLL oncoprotein levels influence leukemia lineage identities.
- Author
-
Janssens DH, Duran M, Otto DJ, Wu W, Xu Y, Kirkey D, Mullighan CG, Yi JS, Meshinchi S, Sarthy JF, Ahmad K, and Henikoff S
- Subjects
- Humans, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Gene Expression Regulation, Leukemic, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins genetics, Mutation, Translocation, Genetic, Hematopoietic Stem Cells metabolism, Chromatin metabolism, Myeloid-Lymphoid Leukemia Protein metabolism, Myeloid-Lymphoid Leukemia Protein genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Oncogene Proteins, Fusion metabolism, Oncogene Proteins, Fusion genetics, Cell Lineage genetics, Histone-Lysine N-Methyltransferase metabolism, Histone-Lysine N-Methyltransferase genetics
- Abstract
Chromosomal translocations involving the mixed-lineage leukemia (MLL) locus generate potent oncogenic fusion proteins (oncoproteins) that disrupt regulation of developmental gene expression. By profiling the oncoprotein-target sites of 36 broadly representative MLL-rearranged leukemia samples, including three samples that underwent a lymphoid-to-myeloid lineage-switching event in response to therapy, we find the genomic enrichment of the oncoprotein is highly variable between samples and subject to dynamic regulation. At high levels of expression, the oncoproteins preferentially activate either an acute lymphoblastic leukemia (ALL) program, enriched for pro-B-cell genes, or an acute myeloid leukemia (AML) program, enriched for hematopoietic-stem-cell genes. The fusion-partner-specific-binding patterns over these gene sets are highly correlated with the prevalence of each mutation in ALL versus AML. In lineage-switching samples the oncoprotein levels are reduced and the oncoproteins preferentially activate granulocyte-monocyte progenitor (GMP) genes. In a sample that lineage switched during treatment with the menin inhibitor revumenib, the oncoprotein and menin are reduced to undetectable levels, but ENL, a transcriptional cofactor of the oncoprotein, persists on numerous oncoprotein-target loci, including genes in the GMP-like lineage-switching program. We propose MLL oncoproteins promote lineage-switching events through dynamic chromatin binding at lineage-specific target genes, and may support resistance to menin inhibitors through similar changes in chromatin occupancy., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
34. Impact of Age on Pharmacogenomics and Treatment Outcomes of B-Cell Acute Lymphoblastic Leukemia.
- Author
-
Yoshimura S, Li Z, Gocho Y, Yang W, Crews KR, Lee SHR, Roberts KG, Mullighan CG, Relling MV, Yu J, Yeoh AEJ, Loh ML, Saygin C, Litzow MR, Jeha S, Karol SE, Inaba H, Pui CH, Konopleva M, Jain N, Stock W, Paietta E, Jabbour E, Kornblau SM, Evans WE, and Yang JJ
- Subjects
- Humans, Adolescent, Child, Adult, Age Factors, Female, Young Adult, Male, Treatment Outcome, Child, Preschool, Aged, Middle Aged, Pharmacogenetics, Antineoplastic Agents therapeutic use, Infant, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Purpose: Acute lymphoblastic leukemia (ALL) can occur across all age groups, with a strikingly higher cure rate in children compared with adults. However, the pharmacological basis of age-related differences in ALL treatment response remains unclear., Methods: Studying 767 children and 309 adults with newly diagnosed B-cell ALL enrolled on frontline trials at St Jude Children's Research Hospital, MD Anderson Cancer Center, the Alliance for Clinical Trials in Oncology, and the ECOG-ACRIN Cancer Research Group, we determined the ex vivo sensitivity of leukemia cells to 21 drugs. Twenty-three ALL molecular subtypes were identified using RNA sequencing. We systematically characterized the associations between drug response and ALL genomics in children, adolescents and young adults, and elderly adults. We evaluated the effect of age-related gene expression signature on ALL treatment outcomes., Results: Seven ALL drugs (asparaginase, prednisolone, mercaptopurine, dasatinib, nelarabine, daunorubicin, and inotuzumab ozogamicin) showed differential activity between children and adults, of which six were explained by age-related differences in leukemia molecular subtypes. Adolescents and young adults showed similar patterns of drug resistance as older adults, relative to young children. Mercaptopurine exhibited subtype-independent greater sensitivity in children. Transcriptomic profiling uncovered subclusters within CRLF2 -, DUX4 -, and KMT2A -rearranged ALL that were linked to age and cytotoxic drug resistance. In particular, a subset of children had adult-like ALL on the basis of leukemia gene expression patterns across subtypes, despite their chronological age. Resistant to cytotoxic drugs, children with adult-like ALL exhibited poor prognosis in pediatric ALL trials, even after adjusting for age and minimal residual diseases., Conclusion: Our results provide pharmacogenomic insights into age-related disparities in ALL cure rates and identify leukemia prognostic features for treatment individualization across age groups.
- Published
- 2024
- Full Text
- View/download PDF
35. Genomic Determinants of Outcome in Acute Lymphoblastic Leukemia.
- Author
-
Chang TC, Chen W, Qu C, Cheng Z, Hedges D, Elsayed A, Pounds SB, Shago M, Rabin KR, Raetz EA, Devidas M, Cheng C, Angiolillo A, Baviskar P, Borowitz M, Burke MJ, Carroll A, Carroll WL, Chen IM, Harvey R, Heerema N, Iacobucci I, Wang JR, Jeha S, Larsen E, Mattano L, Maloney K, Pui CH, Ramirez NC, Salzer W, Willman C, Winick N, Wood B, Hunger SP, Wu G, Mullighan CG, and Loh ML
- Subjects
- Humans, Child, Male, Female, Case-Control Studies, Child, Preschool, Adolescent, Genomics, Infant, PAX5 Transcription Factor genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Purpose: Although cure rates for childhood acute lymphoblastic leukemia (ALL) exceed 90%, ALL remains a leading cause of cancer death in children. Half of relapses arise in children initially classified with standard-risk (SR) disease., Materials and Methods: To identify genomic determinants of relapse in children with SR ALL, we performed genome and transcriptome sequencing of diagnostic and remission samples of children with SR (n = 1,381) or high-risk B-ALL with favorable cytogenetic features (n = 115) enrolled on Children's Oncology Group trials. We used a case-control study design analyzing 439 patients who relapsed and 1,057 who remained in complete remission for at least 5 years., Results: Genomic subtype was associated with relapse, which occurred in approximately 50% of cases of PAX5 -altered ALL (odds ratio [OR], 3.31 [95% CI, 2.17 to 5.03]; P = 3.18 × 10
-8 ). Within high-hyperdiploid ALL, gain of chromosome 10 with disomy of chromosome 7 was associated with favorable outcome (OR, 0.27 [95% CI, 0.17 to 0.42]; P = 8.02 × 10-10 ; St Jude Children's Research Hospital validation cohort: OR, 0.22 [95% CI, 0.05 to 0.80]; P = .009), and disomy of chromosomes 10 and 17 with gain of chromosome 6 was associated with relapse (OR, 7.16 [95% CI, 2.63 to 21.51]; P = 2.19 × 10-5 ; validation cohort: OR, 21.32 [95% CI, 3.62 to 119.30]; P = .0004). Genomic alterations were associated with relapse in a subtype-dependent manner, including alterations of INO80 in ETV6::RUNX1 ALL, IKZF1 , and CREBBP in high-hyperdiploid ALL and FHIT in BCR::ABL1 -like ALL. Genomic alterations were also associated with the presence of minimal residual disease, including NRAS and CREBBP in high-hyperdiploid ALL., Conclusion: Genetic subtype, patterns of aneuploidy, and secondary genomic alterations determine risk of relapse in childhood ALL. Comprehensive genomic analysis is required for optimal risk stratification.- Published
- 2024
- Full Text
- View/download PDF
36. Biologic and Clinical Analysis of Childhood Gamma Delta T-ALL Identifies LMO2/STAG2 Rearrangements as Extremely High Risk.
- Author
-
Kimura S, Park CS, Montefiori LE, Iacobucci I, Pölönen P, Gao Q, Arnold ED, Attarbaschi A, Brown A, Buldini B, Caldwell KJ, Chang Y, Chen C, Cheng C, Cheng Z, Choi J, Conter V, Crews KR, de Groot-Kruseman HA, Deguchi T, Eguchi M, Muhle HE, Elitzur S, Escherich G, Freeman BB 3rd, Gu Z, Han K, Horibe K, Imamura T, Jeha S, Kato M, Chiew KH, Khan T, Kicinski M, Köhrer S, Kornblau SM, Kotecha RS, Li CK, Liu YC, Locatelli F, Luger SM, Paietta EM, Manabe A, Marquart HV, Masetti R, Maybury M, Mazilier P, Meijerink JPP, Mitchell S, Miyamura T, Moore AS, Oshima K, Pawinska-Wasikowska K, Pieters R, Prater MS, Pruett-Miller SM, Pui CH, Qu C, Reiterova M, Reyes N, Roberts KG, Rowe JM, Sato A, Schmiegelow K, Schrappe M, Shen S, Skoczeń S, Spinelli O, Stary J, Svaton M, Takagi M, Takita J, Tang Y, Teachey DT, Thomas PG, Tomizawa D, Trka J, Varotto E, Vincent TL, Yang JJ, Yeoh AEJ, Zhou Y, Zimmermann M, Inaba H, and Mullighan CG
- Subjects
- Humans, Child, Preschool, Male, Female, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Infant, Child, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Gene Rearrangement, Proto-Oncogene Proteins, LIM Domain Proteins genetics, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism
- Abstract
Acute lymphoblastic leukemia expressing the gamma delta T-cell receptor (γδ T-ALL) is a poorly understood disease. We studied 200 children with γδ T-ALL from 13 clinical study groups to understand the clinical and genetic features of this disease. We found age and genetic drivers were significantly associated with outcome. γδ T-ALL diagnosed in children under 3 years of age was extremely high-risk and enriched for genetic alterations that result in both LMO2 activation and STAG2 inactivation. Mechanistically, using patient samples and isogenic cell lines, we show that inactivation of STAG2 profoundly perturbs chromatin organization by altering enhancer-promoter looping, resulting in deregulation of gene expression associated with T-cell differentiation. High-throughput drug screening identified a vulnerability in DNA repair pathways arising from STAG2 inactivation, which can be targeted by poly(ADP-ribose) polymerase inhibition. These data provide a diagnostic framework for classification and risk stratification of pediatric γδ T-ALL. Significance: Patients with acute lymphoblastic leukemia expressing the gamma delta T-cell receptor under 3 years old or measurable residual disease ≥1% at end of induction showed dismal outcomes and should be classified as having high-risk disease. The STAG2/LMO2 subtype was enriched in this very young age group. STAG2 inactivation may perturb chromatin conformation and cell differentiation and confer vulnerability to poly(ADP-ribose) polymerase inhibition., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
37. Classification and risk stratification in T-lineage acute lymphoblastic leukemia.
- Author
-
Pölönen P, Mullighan CG, and Teachey DT
- Abstract
Cure rates for patients with acute lymphoblastic leukemia (ALL) have improved markedly in recent decades, in part due to risk stratification incorporating leukemia genomics, response to treatment, and clinical features to be able to determine at diagnosis which patients are more likely to relapse or have refractory disease. While risk stratification is well-developed for patients with B lineage ALL (B-ALL), it remains challenging for those with T lineage ALL (T-ALL). Prognostic factors validated across clinical trials and real-world data in T-ALL include age, central nervous system (CNS) involvement, and minimal residual disease (MRD) response. Immunophenotype, including early T-cell precursor (ETP) ALL is widely used to classify T-ALL, but is not consistently associated with outcome in multivariable risk models. Historically, few genetic alterations have been consistently associated with outcome, but recent comprehensive, large-scale genomic profiling has identified multiple genetic subtypes and alterations associated with outcome independent of MRD. This review highlights ongoing efforts to identify reliable prognostic biomarkers and underscores the potential of genomics-based classification to guide future T-ALL treatment strategies., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
38. Outcomes in patients with ETV6::RUNX1 or high-hyperdiploid B-ALL treated in the St. Jude Total Therapy XV/XVI studies.
- Author
-
Purvis K, Zhou Y, Karol SE, Rubnitz JE, Ribeiro RC, Lee SH, Yang JJ, Bowman WP, Wang L, Dixon SB, Roberts KG, Gao Q, Cheng C, Mullighan CG, Jeha S, Pui CH, and Inaba H
- Abstract
Children with ETV6::RUNX1 or high-hyperdiploid B-acute lymphoblastic leukemia (B-ALL) have favorable outcomes. The St. Jude (SJ) classification considers these patients low-risk, regardless of their National Cancer Institute (NCI) risk, except when there is slow minimal residual disease (MRD) response or central nervous system/testicular involvement. We analyzed outcomes in children (aged 1-18.99 years) with these genotypes in the SJ Total XV and XVI studies (2000-2017). Patients with ETV6::RUNX1 (n = 222) or high-hyperdiploid (n = 296) B-ALL had 5-year event-free survival (EFS) of 97.7% ± 1.1% and 94.7% ± 1.4%, respectively. For ETV6::RUNX1, EFS was comparable for NCI standard-risk and high-risk patients (97.8% ± 1.2% and 97.5% ± 2.6%, respectively; P = 0.917) and for SJ low-risk and standard-risk patients (97.4% ± 1.2% and 100.0%; P = 0.360). Thirty-seven of 40 NCI high-risk patients who received SJ low-risk therapy had excellent EFS (97.3% ± 2.8%). For high-hyperdiploid B-ALL, EFS was worse for NCI high-risk patients than for standard-risk patients (87.6% ± 4.5% and 96.4% ± 1.3%; P = 0.016). EFS was similar for NCI standard-risk and high-risk patients classified as SJ low-risk (96.0% ± 1.5% and 96.9% ± 3.2%; P = 0.719); however, EFS was worse for NCI high-risk patients than for NCI standard-risk patients receiving SJ standard/high-risk therapy (77.4% ± 8.2% and 98.0% ± 2.2%; P = 0.004). NCI high-risk patients with ETV6::RUNX1 or high-hyperdiploid B-ALL who received SJ low-risk therapy had lower incidences of thrombosis (P = 0.013) and pancreatitis (P = 0.011) than those who received SJ standard/high-risk therapy. Contemporary MRD-directed therapy yielded excellent outcomes, except for NCI high-risk high-hyperdiploid B-ALL patients with slow MRD response, who require new treatment approaches. Among NCI high-risk patients, 93% with ETV6::RUNX1 and 54% with high-hyperdiploid B-ALL experienced excellent outcomes with a low-intensity regimen. These trials were registered at www.clinicaltrials.gov as #NCT00137111 and #NCT00549848., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
39. KMT2A oncoproteins induce epigenetic resistance to targeted therapies.
- Author
-
Janssens DH, Duran M, Otto DJ, Wu W, Xu Y, Kirkey D, Mullighan CG, Yi JS, Meshinchi S, Sarthy JF, Ahmad K, and Henikoff S
- Abstract
Chromosomal translocations involving the Lysine-Methyl-Transferase-2A ( KMT2A ) locus generate potent oncogenic fusion proteins (oncoproteins) that disrupt regulation of developmental gene expression. By profiling the oncoprotein-target sites of 36 broadly representative KMT2A -rearranged leukemia samples, including three samples that underwent a lymphoid-to-myeloid lineage-switching event in response to therapy, we find the genomic enrichment of the oncoprotein is highly variable between samples and subject to dynamic regulation. At high levels of expression, the oncoproteins preferentially activate either an acute lymphoblastic leukemia (ALL) program, enriched for pro-B-cell genes, or an acute myeloid leukemia (AML) program, enriched for hematopoietic-stem-cell genes. The fusion-partner-specific-binding patterns over these gene sets are highly correlated with the prevalence of each mutation in ALL versus AML. In lineage-switching samples the oncoprotein levels are reduced and the oncoproteins preferentially activate granulocyte-monocyte progenitor (GMP) genes. In a sample that lineage switched during treatment with the menin inhibitor revumenib, the oncoprotein and menin are reduced to undetectable levels, but ENL, a transcriptional cofactor of the oncoprotein, persists on numerous oncoprotein-target loci, including genes in the GMP-like lineage-switching program. We propose KMT2A oncoproteins promote lineage-switching events through dynamic chromatin binding and can induce epigenetic lesions, marked by ENL, that support resistance to targeted therapies.
- Published
- 2024
- Full Text
- View/download PDF
40. Bootstrap Evaluation of Association Matrices (BEAM) for Integrating Multiple Omics Profiles with Multiple Outcomes.
- Author
-
Seffernick AE, Cao X, Cheng C, Yang W, Autry RJ, Yang JJ, Pui CH, Teachey DT, Lamba JK, Mullighan CG, and Pounds SB
- Abstract
Motivation: Large datasets containing multiple clinical and omics measurements for each subject motivate the development of new statistical methods to integrate these data to advance scientific discovery., Model: We propose bootstrap evaluation of association matrices (BEAM), which integrates multiple omics profiles with multiple clinical endpoints. BEAM associates a set omic features with clinical endpoints via regression models and then uses bootstrap resampling to determine statistical significance of the set. Unlike existing methods, BEAM uniquely accommodates an arbitrary number of omic profiles and endpoints., Results: In simulations, BEAM performed similarly to the theoretically best simple test and outperformed other integrated analysis methods. In an example pediatric leukemia application, BEAM identified several genes with biological relevance established by a CRISPR assay that had been missed by univariate screens and other integrated analysis methods. Thus, BEAM is a powerful, flexible, and robust tool to identify genes for further laboratory and/or clinical research evaluation., Availability: Source code, documentation, and a vignette for BEAM are available on GitHub at: https://github.com/annaSeffernick/BEAMR. The R package is available from CRAN at: https://cran.r-project.org/package=BEAMR., Contact: Stanley.Pounds@stjude.org., Supplementary Information: Supplementary data are available at the journal's website.
- Published
- 2024
- Full Text
- View/download PDF
41. Conserved epigenetic hallmarks of T cell aging during immunity and malignancy.
- Author
-
Mi T, Soerens AG, Alli S, Kang TG, Vasandan AB, Wang Z, Vezys V, Kimura S, Iacobucci I, Baylin SB, Jones PA, Hiner C, Mueller A, Goldstein H, Mullighan CG, Zebley CC, Masopust D, and Youngblood B
- Subjects
- Animals, Humans, Mice, Aging immunology, Aging genetics, T-Lymphocytes immunology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Memory T Cells immunology, Memory T Cells metabolism, Mice, Inbred C57BL, Male, T-Cell Senescence, Epigenesis, Genetic, Cellular Senescence genetics, Cellular Senescence immunology
- Abstract
Chronological aging correlates with epigenetic modifications at specific loci, calibrated to species lifespan. Such 'epigenetic clocks' appear conserved among mammals, but whether they are cell autonomous and restricted by maximal organismal lifespan remains unknown. We used a multilifetime murine model of repeat vaccination and memory T cell transplantation to test whether epigenetic aging tracks with cellular replication and if such clocks continue 'counting' beyond species lifespan. Here we found that memory T cell epigenetic clocks tick independently of host age and continue through four lifetimes. Instead of recording chronological time, T cells recorded proliferative experience through modification of cell cycle regulatory genes. Applying this epigenetic profile across a range of human T cell contexts, we found that naive T cells appeared 'young' regardless of organism age, while in pediatric patients, T cell acute lymphoblastic leukemia appeared to have epigenetically aged for up to 200 years. Thus, T cell epigenetic clocks measure replicative history and can continue to accumulate well-beyond organismal lifespan., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
42. The genomic basis of childhood T-lineage acute lymphoblastic leukaemia.
- Author
-
Pölönen P, Di Giacomo D, Seffernick AE, Elsayed A, Kimura S, Benini F, Montefiori LE, Wood BL, Xu J, Chen C, Cheng Z, Newman H, Myers J, Iacobucci I, Li E, Sussman J, Hedges D, Hui Y, Diorio C, Uppuluri L, Frank D, Fan Y, Chang Y, Meshinchi S, Ries R, Shraim R, Li A, Bernt KM, Devidas M, Winter SS, Dunsmore KP, Inaba H, Carroll WL, Ramirez NC, Phillips AH, Kriwacki RW, Yang JJ, Vincent TL, Zhao Y, Ghate PS, Wang J, Reilly C, Zhou X, Sanders MA, Takita J, Kato M, Takasugi N, Chang BH, Press RD, Loh M, Rampersaud E, Raetz E, Hunger SP, Tan K, Chang TC, Wu G, Pounds SB, Mullighan CG, and Teachey DT
- Subjects
- Child, Female, Humans, Male, Chromatin genetics, Chromatin metabolism, Enhancer Elements, Genetic genetics, Epigenomics, Gene Expression Regulation, Leukemic, Single-Cell Analysis, Transcriptome genetics, T-Lymphocytes cytology, T-Lymphocytes pathology, Genome, Human genetics, Genomics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
T-lineage acute lymphoblastic leukaemia (T-ALL) is a high-risk tumour
1 that has eluded comprehensive genomic characterization, which is partly due to the high frequency of noncoding genomic alterations that result in oncogene deregulation2,3 . Here we report an integrated analysis of genome and transcriptome sequencing of tumour and remission samples from more than 1,300 uniformly treated children with T-ALL, coupled with epigenomic and single-cell analyses of malignant and normal T cell precursors. This approach identified 15 subtypes with distinct genomic drivers, gene expression patterns, developmental states and outcomes. Analyses of chromatin topology revealed multiple mechanisms of enhancer deregulation that involve enhancers and genes in a subtype-specific manner, thereby demonstrating widespread involvement of the noncoding genome. We show that the immunophenotypically described, high-risk entity of early T cell precursor ALL is superseded by a broader category of 'early T cell precursor-like' leukaemia. This category has a variable immunophenotype and diverse genomic alterations of a core set of genes that encode regulators of hematopoietic stem cell development. Using multivariable outcome models, we show that genetic subtypes, driver and concomitant genetic alterations independently predict treatment failure and survival. These findings provide a roadmap for the classification, risk stratification and mechanistic understanding of this disease., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
43. Blinatumomab for MRD-Negative Acute Lymphoblastic Leukemia in Adults.
- Author
-
Litzow MR, Sun Z, Mattison RJ, Paietta EM, Roberts KG, Zhang Y, Racevskis J, Lazarus HM, Rowe JM, Arber DA, Wieduwilt MJ, Liedtke M, Bergeron J, Wood BL, Zhao Y, Wu G, Chang TC, Zhang W, Pratz KW, Dinner SN, Frey N, Gore SD, Bhatnagar B, Atallah EL, Uy GL, Jeyakumar D, Lin TL, Willman CL, DeAngelo DJ, Patel SB, Elliott MA, Advani AS, Tzachanis D, Vachhani P, Bhave RR, Sharon E, Little RF, Erba HP, Stone RM, Luger SM, Mullighan CG, and Tallman MS
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Antibodies, Bispecific adverse effects, Antibodies, Bispecific therapeutic use, Antibodies, Bispecific administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Consolidation Chemotherapy, Disease-Free Survival, Induction Chemotherapy, Kaplan-Meier Estimate, Recurrence, Remission Induction, Survival Analysis, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, Neoplasm, Residual, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
Background: Many older adults with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) have a relapse despite having a measurable residual disease (MRD)-negative complete remission with combination chemotherapy. The addition of blinatumomab, a bispecific T-cell engager molecule that is approved for the treatment of relapsed, refractory, and MRD-positive BCP-ALL, may have efficacy in patients with MRD-negative remission., Methods: In a phase 3 trial, we randomly assigned patients 30 to 70 years of age with BCR::ABL1 -negative BCP-ALL (with :: indicating fusion) who had MRD-negative remission (defined as <0.01% leukemic cells in bone marrow as assessed on flow cytometry) after induction and intensification chemotherapy to receive four cycles of blinatumomab in addition to four cycles of consolidation chemotherapy or to receive four cycles of consolidation chemotherapy alone. The primary end point was overall survival, and relapse-free survival was a secondary end point., Results: The data and safety monitoring committee reviewed the results from the third efficacy interim analysis and recommended that they be reported. Complete remission with or without full count recovery was observed in 395 of 488 enrolled patients (81%). Of the 224 patients with MRD-negative status, 112 were assigned to each group. The characteristics of the patients were balanced between the groups. At a median follow-up of 43 months, an advantage was observed in the blinatumomab group as compared with the chemotherapy-only group with regard to overall survival (at 3 years: 85% vs. 68%; hazard ratio for death, 0.41; 95% confidence interval [CI], 0.23 to 0.73; P = 0.002), and the 3-year relapse-free survival was 80% with blinatumomab and 64% with chemotherapy alone (hazard ratio for relapse or death, 0.53; 95% CI, 0.32 to 0.87). A higher incidence of neuropsychiatric events was reported in the blinatumomab group than in the chemotherapy-only group., Conclusions: The addition of blinatumomab to consolidation chemotherapy in adult patients in MRD-negative remission from BCP-ALL significantly improved overall survival. (Funded by the National Institutes of Health and others; E1910 ClinicalTrials.gov number, NCT02003222.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
44. Genomic determinants of response and resistance to inotuzumab ozogamicin in B-cell ALL.
- Author
-
Zhao Y, Short NJ, Kantarjian HM, Chang TC, Ghate PS, Qu C, Macaron W, Jain N, Thakral B, Phillips AH, Khoury J, Garcia-Manero G, Zhang W, Fan Y, Yang H, Garris RS, Nasr LF, Kriwacki RW, Roberts KG, Konopleva M, Jabbour EJ, and Mullighan CG
- Subjects
- Humans, Female, Mutation, Male, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Agents, Immunological pharmacology, Adult, Middle Aged, Retrospective Studies, Adolescent, Inotuzumab Ozogamicin, Sialic Acid Binding Ig-like Lectin 2 genetics, Drug Resistance, Neoplasm genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
Abstract: Inotuzumab ozogamicin (InO) is an antibody-drug conjugate that delivers calicheamicin to CD22-expressing cells. In a retrospective cohort of InO-treated patients with B-cell acute lymphoblastic leukemia, we sought to understand the genomic determinants of the response and resistance to InO. Pre- and post-InO-treated patient samples were analyzed by whole genome, exome, and/or transcriptome sequencing. Acquired CD22 mutations were observed in 11% (3/27) of post-InO-relapsed tumor samples, but not in refractory samples (0/16). There were multiple CD22 mutations per sample and the mechanisms of CD22 escape included epitope loss (protein truncation and destabilization) and epitope alteration. Two CD22 mutant cases were post-InO hyper-mutators resulting from error-prone DNA damage repair (nonhomologous/alternative end-joining repair, or mismatch repair deficiency), suggesting that hypermutation drove escape from CD22-directed therapy. CD22-mutant relapses occurred after InO and subsequent hematopoietic stem cell transplantation (HSCT), suggesting that InO eliminated the predominant clones, leaving subclones with acquired CD22 mutations that conferred resistance to InO and subsequently expanded. Acquired loss-of-function mutations in TP53, ATM, and CDKN2A were observed, consistent with a compromise of the G1/S DNA damage checkpoint as a mechanism for evading InO-induced apoptosis. Genome-wide CRISPR/Cas9 screening of cell lines identified DNTT (terminal deoxynucleotidyl transferase) loss as a marker of InO resistance. In conclusion, genetic alterations modulating CD22 expression and DNA damage response influence InO efficacy. Our findings highlight the importance of defining the basis of CD22 escape and eradication of residual disease before HSCT. The identified mechanisms of escape from CD22-targeted therapy extend beyond antigen loss and provide opportunities to improve therapeutic approaches and overcome resistance. These trials were registered at www.ClinicalTrials.gov as NCT01134575, NCT01371630, and NCT03441061., (© 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
45. Perinatal thymic-derived CD8αβ-expressing γδ T cells are innate IFN-γ producers that expand in IL-7R-STAT5B-driven neoplasms.
- Author
-
Sumaria N, Fiala GJ, Inácio D, Curado-Avelar M, Cachucho A, Pinheiro R, Wiesheu R, Kimura S, Courtois L, Blankenhaus B, Darrigues J, Suske T, Almeida ARM, Minguet S, Asnafi V, Lhermitte L, Mullighan CG, Coffelt SB, Moriggl R, Barata JT, Pennington DJ, and Silva-Santos B
- Subjects
- Animals, Mice, Humans, Signal Transduction immunology, Mice, Inbred C57BL, CD8-Positive T-Lymphocytes immunology, Mice, Knockout, Receptors, Antigen, T-Cell, alpha-beta metabolism, Receptors, Antigen, T-Cell, alpha-beta genetics, CD8 Antigens metabolism, Female, Intraepithelial Lymphocytes immunology, Intraepithelial Lymphocytes metabolism, Interleukin-7 metabolism, Interferon-gamma metabolism, Interferon-gamma immunology, Receptors, Antigen, T-Cell, gamma-delta metabolism, Receptors, Antigen, T-Cell, gamma-delta immunology, Thymus Gland immunology, Receptors, Interleukin-7 metabolism, Immunity, Innate, STAT5 Transcription Factor metabolism
- Abstract
The contribution of γδ T cells to immune responses is associated with rapid secretion of interferon-γ (IFN-γ). Here, we show a perinatal thymic wave of innate IFN-γ-producing γδ T cells that express CD8αβ heterodimers and expand in preclinical models of infection and cancer. Optimal CD8αβ
+ γδ T cell development is directed by low T cell receptor signaling and through provision of interleukin (IL)-4 and IL-7. This population is pathologically relevant as overactive, or constitutive, IL-7R-STAT5B signaling promotes a supraphysiological accumulation of CD8αβ+ γδ T cells in the thymus and peripheral lymphoid organs in two mouse models of T cell neoplasia. Likewise, CD8αβ+ γδ T cells define a distinct subset of human T cell acute lymphoblastic leukemia pediatric patients. This work characterizes the normal and malignant development of CD8αβ+ γδ T cells that are enriched in early life and contribute to innate IFN-γ responses to infection and cancer., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
46. Association of leukemic molecular profile with efficacy of inotuzumab ozogamicin in adults with relapsed/refractory ALL.
- Author
-
Zhao Y, Laird AD, Roberts KG, Yafawi R, Kantarjian H, DeAngelo DJ, Stelljes M, Liedtke M, Stock W, Gökbuget N, O'Brien S, Jabbour E, Cassaday RD, Loyd MR, Olsen S, Neale G, Liu X, Vandendries E, Advani A, and Mullighan CG
- Subjects
- Humans, Adult, Female, Male, Middle Aged, Treatment Outcome, Aged, Recurrence, Antineoplastic Agents, Immunological therapeutic use, Young Adult, Drug Resistance, Neoplasm, Adolescent, Inotuzumab Ozogamicin therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality
- Abstract
Abstract: The phase 3 INO-VATE trial demonstrated higher rates of remission, measurable residual disease negativity, and improved overall survival for patients with relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL) who received inotuzumab ozogamicin (InO) vs standard-of-care chemotherapy (SC). Here, we examined associations between genomic alterations and the efficacy of InO. Of 326 randomized patients, 91 (InO, n = 43; SC, n = 48) had samples evaluable for genomic analysis. The spectrum of gene fusions and other genomic alterations observed was comparable with prior studies of adult ALL. Responses to InO were observed in all leukemic subtypes, genomic alterations, and risk groups. Significantly higher rates of complete remission (CR)/CR with incomplete count recovery were observed with InO vs SC in patients with BCR::ABL1-like ALL (85.7% [6/7] vs 0% [0/5]; P = .0076), with TP53 alterations (100% [5/5] vs 12.5% [1/8]; P = .0047), and in the high-risk BCR::ABL1- (BCR::ABL1-like, low-hypodiploid, KMT2A-rearranged) group (83.3% [10/12] vs 10.5% [2/19]; P < .0001). This retrospective, exploratory analysis of the INO-VATE trial demonstrated potential for benefit with InO for patients with R/R ALL across leukemic subtypes, including BCR::ABL1-like ALL, and for those bearing diverse genomic alterations. Further confirmation of the efficacy of InO in patients with R/R ALL exhibiting the BCR::ABL1-like subtype or harboring TP53 alterations is warranted. This trial was registered at www.ClinicalTrials.gov as #NCT01564784., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
47. A lineage-specific STAT5BN642H mouse model to study NK-cell leukemia.
- Author
-
Klein K, Kollmann S, Hiesinger A, List J, Kendler J, Klampfl T, Rhandawa M, Trifinopoulos J, Maurer B, Grausenburger R, Betram CA, Moriggl R, Rülicke T, Mullighan CG, Witalisz-Siepracka A, Walter W, Hoermann G, Sexl V, and Gotthardt D
- Subjects
- Animals, Mice, Humans, Disease Models, Animal, Cell Lineage genetics, Mutation, Mice, Transgenic, STAT5 Transcription Factor genetics, STAT5 Transcription Factor metabolism, Killer Cells, Natural metabolism, Killer Cells, Natural immunology, Killer Cells, Natural pathology, Leukemia, Large Granular Lymphocytic genetics, Leukemia, Large Granular Lymphocytic pathology
- Abstract
Abstract: Patients with T- and natural killer (NK)-cell neoplasms frequently have somatic STAT5B gain-of-function mutations. The most frequent STAT5B mutation is STAT5BN642H, which is known to drive murine T-cell leukemia, although its role in NK-cell malignancies is unclear. Introduction of the STAT5BN642H mutation into human NK-cell lines enhances their potential to induce leukemia in mice. We have generated a mouse model that enables tissue-specific expression of STAT5BN642H and have selectively expressed the mutated STAT5B in hematopoietic cells (N642Hvav/+) or exclusively in NK cells (N642HNK/NK). All N642Hvav/+ mice rapidly develop an aggressive T/NKT-cell leukemia, whereas N642HNK/NK mice display an indolent NK-large granular lymphocytic leukemia (NK-LGLL) that progresses to an aggressive leukemia with age. Samples from patients with NK-cell leukemia have a distinctive transcriptional signature driven by mutant STAT5B, which overlaps with that of murine leukemic N642HNK/NK NK cells. To our knowledge, we have generated the first reliable STAT5BN642H-driven preclinical mouse model that displays an indolent NK-LGLL progressing to aggressive NK-cell leukemia. This novel in vivo tool will enable us to explore the transition from an indolent to an aggressive disease and will thus permit the study of prevention and treatment options for NK-cell malignancies., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
48. Acute lymphoblastic leukaemia.
- Author
-
Pagliaro L, Chen SJ, Herranz D, Mecucci C, Harrison CJ, Mullighan CG, Zhang M, Chen Z, Boissel N, Winter SS, and Roti G
- Subjects
- Humans, Genomics, Molecular Targeted Therapy, Quality of Life, Immunotherapy, Adoptive, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma physiopathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Acute lymphoblastic leukaemia (ALL) is a haematological malignancy characterized by the uncontrolled proliferation of immature lymphoid cells. Over past decades, significant progress has been made in understanding the biology of ALL, resulting in remarkable improvements in its diagnosis, treatment and monitoring. Since the advent of chemotherapy, ALL has been the platform to test for innovative approaches applicable to cancer in general. For example, the advent of omics medicine has led to a deeper understanding of the molecular and genetic features that underpin ALL. Innovations in genomic profiling techniques have identified specific genetic alterations and mutations that drive ALL, inspiring new therapies. Targeted agents, such as tyrosine kinase inhibitors and immunotherapies, have shown promising results in subgroups of patients while minimizing adverse effects. Furthermore, the development of chimeric antigen receptor T cell therapy represents a breakthrough in ALL treatment, resulting in remarkable responses and potential long-term remissions. Advances are not limited to treatment modalities alone. Measurable residual disease monitoring and ex vivo drug response profiling screening have provided earlier detection of disease relapse and identification of exceptional responders, enabling clinicians to adjust treatment strategies for individual patients. Decades of supportive and prophylactic care have improved the management of treatment-related complications, enhancing the quality of life for patients with ALL., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
49. CASZ1 upregulates PI3K-AKT-mTOR signaling and promotes T-cell acute lymphoblastic leukemia.
- Author
-
Cardoso BA, Duque M, Gírio A, Fragoso R, Oliveira ML, Allen JR, Martins LR, Correia NC, Silveira AB, Veloso A, Kimura S, Demoen L, Matthijssens F, Jeha S, Cheng C, Pui CH, Grosso AR, Neto JL, De Almeida SF, Van Vlieberghe P, Mullighan CG, Yunes JA, Langenau DM, Pflumio F, and Barata JT
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Gene Expression Regulation, Leukemic, Receptor, Notch1 metabolism, Receptor, Notch1 genetics, Transcription Factors genetics, Transcription Factors metabolism, Phosphatidylinositol 3-Kinases metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, T-Cell Acute Lymphocytic Leukemia Protein 1 metabolism, T-Cell Acute Lymphocytic Leukemia Protein 1 genetics, TOR Serine-Threonine Kinases metabolism, Zebrafish
- Abstract
CASZ1 is a conserved transcription factor involved in neural development, blood vessel assembly and heart morphogenesis. CASZ1 has been implicated in cancer, either suppressing or promoting tumor development depending on the tissue. However, the impact of CASZ1 on hematological tumors remains unknown. Here, we show that the T-cell oncogenic transcription factor TAL1 is a direct positive regulator of CASZ1, that T-cell acute lymphoblastic leukemia (T-ALL) samples at diagnosis overexpress CASZ1b isoform, and that CASZ1b expression in patient samples correlates with PI3K-AKT-mTOR signaling pathway activation. In agreement, overexpression of CASZ1b in both Ba/F3 and T-ALL cells leads to the activation of PI3K signaling pathway, which is required for CASZ1b-mediated transformation of Ba/F3 cells in vitro and malignant expansion in vivo. We further demonstrate that CASZ1b cooperates with activated NOTCH1 to promote T-ALL development in zebrafish, and that CASZ1b protects human T-ALL cells from serum deprivation and treatment with chemotherapeutic drugs. Taken together, our studies indicate that CASZ1b is a TAL1-regulated gene that promotes T-ALL development and resistance to chemotherapy.
- Published
- 2024
- Full Text
- View/download PDF
50. Proximally biased V(D)J recombination in the clonal evolution of IGH alleles in KMT2A::AFF1 BCP-ALL of all age classes.
- Author
-
Müller H, Dicker F, Bär C, Walter W, Hutter S, Nadarajah N, Meggendorfer M, Gao Q, Iacobucci I, Mullighan CG, Kern W, Haferlach T, and Haferlach C
- Abstract
Competing Interests: Torsten Haferlach, Claudia Haferlach, and Wolfgang Kern declare part ownership of Munich Leukemia Laboratory (MLL). Heiko Müller, Frank Dicker, Constance Bär, Wencke Walter, Stephan Hutter, Niroshan Nadarajah, and Manja Meggendorfer are employed by the MLL. Charles G. Mullighan received research funding from Loxo Oncology, Pfizer, AbbVie; honoraria from Amgen and Illumina, and holds stock in Amgen.
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.