202 results on '"Infant Acute Lymphoblastic Leukemia"'
Search Results
2. Immunological assessment of a patient with infant acute lymphoblastic leukemia.
- Author
-
Takagi M, Tomoyasu C, Yamanaka J, Hamabata T, Isoda T, Miyamura T, and Imai K
- Subjects
- Infant, Humans, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma
- Published
- 2024
- Full Text
- View/download PDF
3. Biology and Treatment of Acute Leukemias in Infants : Perspective 2
- Author
-
Reaman, Gregory H., Markman, Maurie, editor, and Pui, Ching-Hon, editor
- Published
- 2003
- Full Text
- View/download PDF
4. Clinical Implications of Minimal Residual Disease Detection in Infants With KMT2A-Rearranged Acute Lymphoblastic Leukemia Treated on the Interfant-06 Protocol
- Author
-
Tomasz Szczepański, Claus Meyer, Maria Grazia Valsecchi, Jan Stary, Birgitte Lausen, Alina Ferster, Gabriele Escherich, Inge M. van der Sluis, Martin Schrappe, Myriam Campbell, Vincent H.J. van der Velden, Giovanni Cazzaniga, Rishi S. Kotecha, Luca Lo Nigro, Julia Alten, Andishe Attarbaschi, Franco Locatelli, Andrea Biondi, Ajay Vora, Rolf Marschalek, Chi Kong Li, Jan Zuna, Benoit Brethon, Rob Pieters, Paola De Lorenzo, Philip Ancliffe, Janine Stutterheim, Stutterheim, J, van der Sluis, I, de Lorenzo, P, Alten, J, Ancliffe, P, Attarbaschi, A, Brethon, B, Biondi, A, Campbell, M, Cazzaniga, G, Escherich, G, Ferster, A, Kotecha, R, Lausen, B, Li, C, Lo Nigro, L, Locatelli, F, Marschalek, R, Meyer, C, Schrappe, M, Stary, J, Vora, A, Zuna, J, van der Velden, V, Szczepanski, T, Valsecchi, M, Pieters, R, and Immunology
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,KMT2A gene ,Lymphoblastic Leukemia ,Incidence (epidemiology) ,medicine.disease ,Minimal residual disease ,Infant Acute Lymphoblastic Leukemia ,Minimal Residual Disease, Infant Acute Lymphoblastic Leukemia, KMT2A ,KMT2A ,MRD ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,SDG 3 - Good Health and Well-being ,Internal medicine ,biology.protein ,Medicine ,Neoplasm ,business ,ALL - Abstract
PURPOSE Infant acute lymphoblastic leukemia (ALL) is characterized by a high incidence of KMT2A gene rearrangements and poor outcome. We evaluated the value of minimal residual disease (MRD) in infants with KMT2A-rearranged ALL treated within the Interfant-06 protocol, which compared lymphoid-style consolidation (protocol IB) versus myeloid-style consolidation (araC, daunorubicin, etoposide/mitoxantrone, araC, etoposide). MATERIALS AND METHODS MRD was measured in 249 infants by DNA-based polymerase chain reaction of rearranged KMT2A, immunoglobulin, and/or T-cell receptor genes, at the end of induction (EOI) and end of consolidation (EOC). MRD results were classified as negative, intermediate (< 5 × 10−4), and high (≥ 5 × 10−4). RESULTS EOI MRD levels predicted outcome with 6-year disease-free survival (DFS) of 60.2% (95% CI, 43.2 to 73.6), 45.0% (95% CI, 28.3 to 53.1), and 33.8% (95% CI, 23.8 to 44.1) for infants with negative, intermediate, and high EOI MRD levels, respectively ( P = .0039). EOC MRD levels were also predictive of outcome, with 6-year DFS of 68.2% (95% CI, 55.2 to 78.1), 40.1% (95% CI, 28.1 to 51.9), and 11.9% (95% CI, 2.6 to 29.1) for infants with negative, intermediate, and high EOC MRD levels, respectively ( P < .0001). Analysis of EOI MRD according to the type of consolidation treatment showed that infants treated with lymphoid-style consolidation had 6-year DFS of 78.2% (95% CI, 51.4 to 91.3), 47.2% (95% CI, 33.0 to 60.1), and 23.2% (95% CI, 12.1 to 36.4) for negative, intermediate, and high MRD levels, respectively ( P < .0001), while for myeloid-style–treated patients the corresponding figures were 45.0% (95% CI, 23.9 to 64.1), 41.3% (95% CI, 23.2 to 58.5), and 45.9% (95% CI, 29.4 to 60.9). CONCLUSION This study provides support for the idea that induction therapy selects patients for subsequent therapy; infants with high EOI MRD may benefit from AML-like consolidation (DFS 45.9% v 23.2%), whereas patients with low EOI MRD may benefit from ALL-like consolidation (DFS 78.2% v 45.0%). Patients with positive EOC MRD had dismal outcomes. These findings will be used for treatment interventions in the next Interfant protocol.
- Published
- 2021
5. Combinatorial efficacy of entospletinib and chemotherapy in patient-derived xenograft models of infant acute lymphoblastic leukemia
- Author
-
Min Wang, Joseph P. Loftus, Lisa M Niswander, Stacey Tannheimer, Patrick A. Brown, Anella Yahiaoui, Sarah K. Tasian, Asen Bagashev, and Allyson Schauf
- Subjects
Vincristine ,Indazoles ,Syk ,Article ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,biology ,business.industry ,MEK inhibitor ,Infant ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,KMT2A ,Pyrazines ,biology.protein ,Selumetinib ,Cancer research ,Heterografts ,business ,Myeloid-Lymphoid Leukemia Protein ,030215 immunology ,medicine.drug - Abstract
Survival of infants with KMT2A-rearranged acute lymphoblastic leukemia (ALL) remains dismal despite intensive chemotherapy. We observed constitutive phosphorylation of spleen tyrosine kinase (SYK) and associated signaling proteins in infant ALL patient-derived xenograft (PDX) model specimens and hypothesized that the SYK inhibitor entospletinib would inhibit signaling and cell growth in vitro and leukemia proliferation in vivo. We further predicted that combined entospletinib and chemotherapy could augment anti-leukemia effects. Basal kinase signaling activation and HOXA9/MEIS1 expression differed among KMT2Arearranged (KMT2A-AFF1 [n=4], KMT2A-MLLT3 [n=1], KMT2A-MLLT1 [n=4]) and non-KMT2A-rearranged [n=3] ALL specimens and stratified by genetic subgroup. Incubation of KMT2A-rearranged ALL cells in vitro with entospletinib inhibited methylcellulose colony formation and SYK pathway signaling in a dose-dependent manner. In vivo inhibition of leukemia proliferation with entospletinib monotherapy was observed in RAS-wild-type KMT2A-AFF1, KMT2A-MLLT3, and KMT2A-MLLT1 ALL PDX models with enhanced activity in combination with vincristine chemotherapy in several models. Surprisingly, entospletinib did not decrease leukemia burden in two KMT2A-AFF1 PDX models with NRAS or KRAS mutations, suggesting potential RAS-mediated resistance to SYK inhibition. As hypothesized, superior inhibition of ALL proliferation was observed in KMT2A-AFF1 PDX models treated with entospletinib and the MEK inhibitor selumetinib versus vehicle or inhibitor monotherapies (P
- Published
- 2020
6. Blinatumomab for infant acute lymphoblastic leukemia
- Author
-
Jack Bartram, Michelle Cummins, Sara Ghorashian, Vasudha Nandagudi Rao, Brenda Gibson, Vesna Pavasovic, P Ancliff, Anupama Rao, Katherine Clesham, Beki James, Andrea Malone, Ajay Vora, Sujith Samarasinghe, Katharine Patrick, David O'Connor, and Denise Bonney
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Immunology ,Antineoplastic Agents ,Biochemistry ,Internal medicine ,Acute lymphocytic leukemia ,Antibodies, Bispecific ,medicine ,Humans ,Retrospective Studies ,business.industry ,Infant, Newborn ,Infant ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,Survival Rate ,Child, Preschool ,Female ,Blinatumomab ,business ,Follow-Up Studies ,medicine.drug - Published
- 2020
- Full Text
- View/download PDF
7. Efficacy of combined CDK9/BET inhibition in preclinical models of MLL-rearranged acute leukemia
- Author
-
John Toubia, Hannah McCalmont, Debora A. Casolari, Chelsea Mayoh, Saumya E. Samaraweera, Richard J D'Andrea, Sarah C Bray, Ka Leung Li, Ian D. Lewis, Rab K. Prinjha, Nicholas Smithers, Luke Jones, Shudong Wang, Richard B. Lock, McCalmont, Hannah, Li, Ka Leung, Jones, Luke, Toubia, John, Bray, Sarah C, Casolari, Debora A, Mayoh, Chelsea, Samaraweera, Saumya E, Lewis, Ian D, Prinjha, Rab K, Smithers, Nicholas, Wang, Shudong, and Lock, Richard B
- Subjects
0301 basic medicine ,BRD4 ,cyclin-dependent kinase inhibitors ,Mice ,03 medical and health sciences ,0302 clinical medicine ,stimulus report ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,lymphoid neoplasia ,P-TEFb ,neoplasms ,Acute leukemia ,biology ,leukemia ,acute ,Myeloid leukemia ,Hematology ,medicine.disease ,Cyclin-Dependent Kinase 9 ,Stimulus Report ,Infant Acute Lymphoblastic Leukemia ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,KMT2A ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Myeloid-Lymphoid Leukemia Protein ,Female ,myeloid neoplasia - Abstract
Chromosomal rearrangements of the lysine methyltransferase 2A (KMT2A or MLL) gene are observed in ;10% of all acute leukemias, with particularly high frequency (;80%) in infant acute lymphoblastic leukemia (ALL),1 where, despite aggressive chemotherapy, patients still experience poor outcome and long-term side effects.2 Mixed lineage leukemia (MLL) rearrangements (MLL-r)also indicate particularly poor outcomes for patients with acute myeloid leukemia (AML).3 Mechanistically, MLL-r frequently generates fusion proteins involving partners that function in the super elongation complex,4 the result of which is aberrant recruitment to MLL target genes of the positive transcription elongation factor b (PTEFb), composed of cyclin-dependent kinase 9 (CDK9)as the catalytic subunit.5 CDK9 positively regulates transcription elongation through phosphorylation of serine 2 of RNA polymerase II (RNAPII).6 Given the central role of CDK9 in the leukemic MLL-r gene-expression program,7 and the well-described ability of CDK9 inhibitors to reduce levelsof the short-lived prosurvival protein MCL1,8 a number of CDK9 inhibitors have been selected forclinical trials focusing on acute leukemias, including those with MLL-r.8,9 In MLL-r leukemia, the bromodomain and extraterminal (BET) family member bromodomain-containing 4 (BRD4)10 acts to recruit PTEFb to super enhancers and together with CDK9 drives increased expression of many oncogenes including MYC.11,12 The roles of CDK9 and BRD4 in MLL-r leukemias present a strong case for testing inhibitors of these proteins in combination as a potential treatment of MLL-r acute leukemias. Refereed/Peer-reviewed
- Published
- 2020
- Full Text
- View/download PDF
8. Real-world use of tisagenlecleucel in infant acute lymphoblastic leukemia
- Author
-
Heather E. Stefanski, Liora M. Schultz, Yasemin Goksenin, Michelle L. Hermiston, Vanessa A Fabrizio, Christina Baggott, Nicole Karras, M. Christa Krupski, Vasant Chinnabhandar, Steven P. Margossian, Holly L Pacenta, Christine L Phillips, Douglas Myers, Kevin J. Curran, Lauren Pommert, Rachel Wilcox, Patrick A. Brown, Muna Qayed, Amy K. Keating, Amy Moskop, Erin H. Breese, Michael R. Verneris, Jenna Rossoff, Cara A Rabik, Crystal L. Mackall, Theodore W. Laetsch, Prakash Satwani, Snehit Prabhu, Julie-An Talano, and Erin M. Guest
- Subjects
Pediatric Research Initiative ,Pediatrics ,medicine.medical_specialty ,Childhood Leukemia ,Pediatric Cancer ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,Vaccine Related ,Rare Diseases ,Clinical Research ,Receptors ,Immunology and Allergy ,Medicine ,Humans ,Antigens ,Child ,Cancer ,Retrospective Studies ,Pediatric ,Transplantation ,Receptors, Chimeric Antigen ,CD19 ,5.2 Cellular and gene therapies ,business.industry ,Chimeric Antigen ,Evaluation of treatments and therapeutic interventions ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,T-Cell ,Stem Cell Research ,United States ,Infant Acute Lymphoblastic Leukemia ,Orphan Drug ,5.1 Pharmaceuticals ,Antigen ,6.1 Pharmaceuticals ,Molecular Medicine ,Immunization ,Development of treatments and therapeutic interventions ,business ,Biotechnology - Abstract
Infants with B-cell acute lymphoblastic leukemia (B-ALL) have poor outcomes because of chemotherapy resistance leading to high relapse rates. Tisagenlecleucel, a CD19-directed chimeric antigen receptor T-cell (CART) therapy, is US Food and Drug Administration approved for relapsed or refractory B-ALL in patients ≤25 years; however, the safety and efficacy of this therapy in young patients is largely unknown because children M1 marrow) were refractory to this therapy (n = 5). Overall, tisagenlecleucel was tolerable in this population, with only 3 patients experiencing ≥grade 3 cytokine release syndrome. No neurotoxicity was reported. This is the largest report of tisagenlecleucel use in infant B-ALL and shows that this therapy is safe and can be effective in this population. Incorporating this novel immunotherapy into the treatment of infant B-ALL offers a promising therapy for a highly aggressive leukemia.
- Published
- 2021
9. Romidepsin enhances the efficacy of cytarabine in vivo, revealing histone deacetylase inhibition as a promising therapeutic strategy for KMT2A-rearranged infant acute lymphoblastic leukemia
- Author
-
Jette Ford, Sébastien Malinge, Joyce Oommen, Sajla Singh, Anastasia M. Hughes, Emanuela Ferrari, Grace-Alyssa Chua, Mark N. Cruickshank, Rishi S. Kotecha, Laurence C. Cheung, Ursula R. Kees, and Richard B. Lock
- Subjects
biology ,business.industry ,Cell growth ,Hematology ,Gene rearrangement ,Infant Acute Lymphoblastic Leukemia ,Romidepsin ,KMT2A ,In vivo ,Cancer research ,Cytarabine ,medicine ,biology.protein ,Histone deacetylase ,business ,medicine.drug - Published
- 2019
- Full Text
- View/download PDF
10. Nuclear FGFR2 Interacts with the MLL-AF4 Oncogenic Chimera and Positively Regulates HOXA9 Gene Expression in t(4;11) Leukemia Cells
- Author
-
Daniela Sarnataro, Fabio Cattaneo, Gabriella Esposito, Tiziana Fioretti, Mariateresa Zanobio, Maddalena Raia, Armando Cevenini, Rosario Ammendola, Fioretti, T., Cevenini, A., Zanobio, M., Raia, M., Sarnataro, D., Cattaneo, F., Ammendola, R., and Esposito, G.
- Subjects
musculoskeletal diseases ,MLL-AF4 ,Oncogene Proteins, Fusion ,QH301-705.5 ,Chromosomal translocation ,Target therapy ,Catalysis ,Translocation, Genetic ,Inorganic Chemistry ,Chimera (genetics) ,T(4 ,Cell Line, Tumor ,hemic and lymphatic diseases ,Gene expression ,medicine ,Nucleu ,Physical and Theoretical Chemistry ,Receptor, Fibroblast Growth Factor, Type 2 ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,Gene knockdown ,cell culture ,AF4 ,integumentary system ,Fibroblast growth factor receptor 2 ,Chemistry ,Organic Chemistry ,nucleus ,Homeodomain Protein ,General Medicine ,HOXA9 ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Computer Science Applications ,Cell biology ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,Haematopoiesis ,stomatognathic diseases ,FGFR2 ,11) leukemia ,embryonic structures ,Fibroblast Growth Factor 2 ,Myeloid-Lymphoid Leukemia Protein ,Human - Abstract
The chromosomal translocation t(4;11) marks an infant acute lymphoblastic leukemia associated with dismal prognosis. This rearrangement leads to the synthesis of the MLL-AF4 chimera, which exerts its oncogenic activity by upregulating transcription of genes involved in hematopoietic differentiation. Crucial for chimera’s aberrant activity is the recruitment of the AF4/ENL/P-TEFb protein complex. Interestingly, a molecular interactor of AF4 is fibroblast growth factor receptor 2 (FGFR2). We herein analyze the role of FGFR2 in the context of leukemia using t(4;11) leukemia cell lines. We revealed the interaction between MLL-AF4 and FGFR2 by immunoprecipitation, western blot, and immunofluorescence experiments; we also tested the effects of FGFR2 knockdown, FGFR2 inhibition, and FGFR2 stimulation on the expression of the main MLL-AF4 target genes, i.e., HOXA9 and MEIS1. Our results show that FGFR2 and MLL-AF4 interact in the nucleus of leukemia cells and that FGFR2 knockdown, which is associated with decreased expression of HOXA9 and MEIS1, impairs the binding of MLL-AF4 to the HOXA9 promoter. We also show that stimulation of leukemia cells with FGF2 increases nuclear level of FGFR2 in its phosphorylated form, as well as HOXA9 and MEIS1 expression. In contrast, preincubation with the ATP-mimetic inhibitor PD173074, before FGF2 stimulation, reduced FGFR2 nuclear amount and HOXA9 and MEIS1 transcript level, thereby indicating that MLL-AF4 aberrant activity depends on the nuclear availability of FGFR2. Overall, our study identifies FGFR2 as a new and promising therapeutic target in t(4;11) leukemia.
- Published
- 2021
11. FLT3 inhibitor lestaurtinib plus chemotherapy for newly diagnosed KMT2A-rearranged infant acute lymphoblastic leukemia: Children’s Oncology Group trial AALL0631
- Author
-
Joanne M. Hilden, Mignon L. Loh, Stephen P. Hunger, Lia Gore, Elizabeth A. Raetz, Naomi J. Winick, William L. Carroll, Donald Small, John A. Kairalla, Cindy Wang, Wanda L. Salzer, Nyla A. Heerema, Zo Ann E. Dreyer, Meenakshi Devidas, Michael J. Borowitz, Andrew J. Carroll, and Patrick A. Brown
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Carbazoles ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Furans ,Protein Kinase Inhibitors ,Chemotherapy ,biology ,business.industry ,Lestaurtinib ,Infant ,Histone-Lysine N-Methyltransferase ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,030104 developmental biology ,KMT2A ,fms-Like Tyrosine Kinase 3 ,030220 oncology & carcinogenesis ,Pharmacodynamics ,biology.protein ,Female ,FLT3 Inhibitor ,business ,Myeloid-Lymphoid Leukemia Protein ,Ex vivo ,medicine.drug - Abstract
Infants with KMT2A‐rearranged acute lymphoblastic leukemia (KMT2A-r ALL) have a poor prognosis. KMT2A-r ALL overexpresses FLT3, and the FLT3 inhibitor (FLT3i) lestaurtinib potentiates chemotherapy‐induced cytotoxicity in preclinical models. Children’s Oncology Group (COG) AALL0631 tested whether adding lestaurtinib to post‐induction chemotherapy improved event-free survival (EFS). After chemotherapy induction, KMT2A-r infants received either chemotherapy only or chemotherapy plus lestaurtinib. Correlative assays included FLT3i plasma pharmacodynamics (PD), which categorized patients as inhibited or uninhibited, and FLT3i ex vivo sensitivity (EVS), which categorized leukemic blasts as sensitive or resistant. There was no difference in 3-year EFS between patients treated with chemotherapy plus lestaurtinib (n = 67, 36 ± 6%) vs. chemotherapy only (n = 54, 39 ± 7%, p = 0.67). However, for the lestaurtinib-treated patients, FLT3i PD and FLT3i EVS significantly correlated with EFS. For FLT3i PD, EFS for inhibited/uninhibited was 59 ± 10%/28 ± 7% (p = 0.009) and for FLTi EVS, EFS for sensitive/resistant was 52 ± 8%/5 ± 5% (p
- Published
- 2021
12. Infant Acute Myeloid Leukemia: A Unique Clinical and Biological Entity
- Author
-
Guy Leverger, Odile Fenneteau, Arnaud Petit, Charlotte Calvo, Francoise Mechinaud, André Baruchel, Hôpital Robert Debré Paris, Hôpital Robert Debré, Service d'hématologie-immunologie-oncologie pédiatrique [CHU Trousseau], CHU Trousseau [APHP], and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,[SDV]Life Sciences [q-bio] ,Review ,acute myeloid leukemia ,lcsh:RC254-282 ,Very frequent ,03 medical and health sciences ,Therapeutic approach ,Acute megakaryoblastic leukemia ,0302 clinical medicine ,Age groups ,AML ,Internal medicine ,hemic and lymphatic diseases ,medicine ,neoplasms ,childhood ,business.industry ,Biological entity ,Myeloid leukemia ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Phenotype ,infant ,3. Good health ,Infant Acute Lymphoblastic Leukemia ,030104 developmental biology ,030220 oncology & carcinogenesis ,business - Abstract
Simple Summary Infant acute myeloid leukemia (AML) is a rare subgroup of AML of pediatric patients under two years of age. For a long time, they have been included in pediatric forms of AML. However, infant AML demonstrates unusual clinical and biological characteristics, and its prognosis differs from AML in older children. Most treatment protocols do not distinguish age subgroups in pediatric AML, when in reality these unusual forms of infancy might benefit from specific targeted therapeutics. Herein we summarize the various specificities of infant AML. Abstract Infant acute myeloid leukemia (AML) is a rare subgroup of AML of children
- Published
- 2021
- Full Text
- View/download PDF
13. WITHDRAWN: Clinical features and outcomes of infant acute lymphoblastic leukemia from a single center in China
- Author
-
Hui Li, Zhuo Wang, Kaili Li, Hao Xiong, Ping Zhou, Fang Tao, Jianxin Li, Zhi Chen, and Yi Li
- Subjects
Pediatrics ,medicine.medical_specialty ,Oncology ,business.industry ,Pediatrics, Perinatology and Child Health ,Medicine ,Hematology ,business ,Single Center ,Infant Acute Lymphoblastic Leukemia - Published
- 2021
- Full Text
- View/download PDF
14. Identification and characterization of relapse-initiating cells in MLL-rearranged infant ALL by single-cell transcriptomics
- Author
-
Luke Jones, Ronald W. Stam, Pauline Schneider, Tito Candelli, Dedeke Rockx-Brouwer, Eduard Bodewes, Frank C. P. Holstege, P. Castro, Thanasis Margaritis, and Rob Pieters
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Cell ,Malignancy ,Article ,03 medical and health sciences ,Basal (phylogenetics) ,0302 clinical medicine ,Prednisone ,Cancer genomics ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Medicine ,Humans ,Child ,Gene Rearrangement ,business.industry ,Gene Expression Regulation, Leukemic ,Infant, Newborn ,Infant ,Hematology ,Histone-Lysine N-Methyltransferase ,Translational research ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Prognosis ,3. Good health ,Infant Acute Lymphoblastic Leukemia ,Survival Rate ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Child, Preschool ,Cancer research ,Female ,Bone marrow ,Neoplasm Recurrence, Local ,Single-Cell Analysis ,business ,Transcriptome ,Glucocorticoid ,Myeloid-Lymphoid Leukemia Protein ,medicine.drug ,Follow-Up Studies - Abstract
Infants with MLL-rearranged infant acute lymphoblastic leukemia (MLL-r iALL) undergo intense therapy to counter a highly aggressive malignancy with survival rates of only 30–40%. The majority of patients initially show therapy response, but in two-thirds of cases the leukemia returns, typically during treatment. The glucocorticoid drug prednisone is established as a major player in the treatment of leukemia and the in vivo response to prednisone monotreatment is currently the best indicator of risk for MLL-r iALL. We used two different single-cell RNA sequencing technologies to analyze the expression of a prednisone-dependent signature, derived from an independent study, in diagnostic bone marrow and peripheral blood biopsies. This allowed us to classify individual leukemic cells as either resistant or sensitive to treatment and show that quantification of these two groups can be used to better predict the occurrence of future relapse in individual patients. This work also sheds light on the nature of the therapy-resistant subpopulation of relapse-initiating cells. Leukemic cells associated with high relapse risk are characterized by basal activation of glucocorticoid response, smaller size, and a quiescent gene expression program with cell stemness properties. These results improve current risk stratification and elucidate leukemic therapy-resistant subpopulations at diagnosis.
- Published
- 2021
- Full Text
- View/download PDF
15. XRCC4 rs28360071 intronic variant is associated with increased risk for infant acute lymphoblastic leukemia with KMT2A rearrangements
- Author
-
Cíntia Barros Santos-Rebouças, Francianne Gomes Andrade, Gisele Dallapicola Brisson, Rodolpho Mattos Albano, Maria S. Pombo-de-Oliveira, Ingrid Sardou Cezar, Ana Rossini, Orlando Louzada-Neto, and Bruno Almeida Lopes
- Subjects
0106 biological sciences ,0301 basic medicine ,Myeloid ,nonhomologous end-joining ,Biology ,QH426-470 ,01 natural sciences ,03 medical and health sciences ,Genotype ,medicine ,Genetics ,Allele ,Risk factor ,Molecular Biology ,XRCC4 ,XRCC6 ,Acute leukemia ,Odds ratio ,KMT2A ,Infant Acute Lymphoblastic Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Human and Medical Genetics ,biology.protein ,Early age acute leukemia ,010606 plant biology & botany - Abstract
Early age acute leukemia (EAL) shows a high frequency of KMT2A-rearrangements (KMT2A-r). Previous investigations highlighted double-strand breaks arising from maternal exposure to xenobiotics during pregnancy as a risk factor for EAL and KMT2A-r. In this case-control study, we investigated the relationship between EAL and genetic variants of the nonhomologous end-joining (XRCC6 rs5751129, XRCC4 rs6869366 and rs28360071), since they might affect DNA repair capacity, leading to KMT2A-r and leukemogenesis. Samples from 577 individuals (acute lymphoblastic leukemia-ALL, n=164; acute myeloid leukemia-AML, n=113; controls, n=300) were genotyped. No significant association was found for rs5751129 and rs6869366, whereas rs28360071 was associated with an increased risk for ALL with KMT2A-r (IIxID: OR - Odds ratio 2.23, CI 1.17-4.25, p=0.014). Bone marrow samples from ALL patients showed a higher expression of XRCC4 compared to AML patients (p=0.025). Human Splicing Finder 3.1 predicted that the deleted allele of rs28360071 is potentially associated with the activation of a 5’ cryptic splice site in intron 3 of XRCC4. The sequencing of cDNA did not show any differences on the splicing process for the rs28360071 genotypes. Our results suggest that the deleted allele for rs28360071 increases the risk for ALL with KMT2A-r, but not by modifying the XRCC4 expression levels or its structure.
- Published
- 2020
16. Preclinical efficacy of gemcitabine in MLL-rearranged infant acute lymphoblastic leukemia
- Author
-
Patricia Garrido Castro, Bianca Koopmans, Priscilla Wander, Susan T.C.J.M. Arentsen-Peters, Jan J. Molenaar, Ronald W. Stam, C. Michel Zwaan, Sandra S. Pinhanҫos, Laurence C. Cheung, Sajla Singh, Rob Pieters, Rishi S. Kotecha, Pauline Schneider, M. Emmy M. Dolman, Mark Kerstjens, Grace Alyssa Chua, Luke Jones, UU BETA RESEARCH, and Pediatrics
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Antimetabolites ,Drug Evaluation, Preclinical ,Antimetabolites, Antineoplastic/pharmacology ,Deoxycytidine/analogs & derivatives ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy ,Gene Rearrangement ,Antineoplastic/pharmacology ,business.industry ,Animal ,Myeloid-Lymphoid Leukemia Protein/genetics ,Antineoplastic Combined Chemotherapy Protocols/adverse effects ,Disease Management ,Infant ,Hematology ,Gemcitabine ,Preclinical ,Infant Acute Lymphoblastic Leukemia ,Disease Models, Animal ,Disease Models ,Drug Evaluation ,business ,medicine.drug - Abstract
The treatment of children diagnosed with acute lymphoblastic leukemia (ALL) has improved significantly over recent decades, with 5-year event-free survival (EFS) approaching 85% [1]. However, 5-year EFS for infants diagnosed at less than 1 year of age with MLL-rearranged ALL remains less than 40% [2]. MLL-rearranged infant ALL is both clinically and biologically distinct from other childhood ALL subtypes, which is reflected by a unique gene expression profile [3], a remarkably silent mutational landscape [4], and pronounced resistance to currently applied chemotherapeutics [5].
- Published
- 2020
17. Chimeric antigen receptor T‐cell therapy for marrow and extramedullary relapse of infant acute lymphoblastic leukemia
- Author
-
Lauren Pommert, Amy Moskop, Pooja D. Thakrar, Julie Talano, and Rachel Phelan
- Subjects
medicine.medical_treatment ,Immunotherapy, Adoptive ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Antigen ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Receptors, Chimeric Antigen ,biology ,business.industry ,Hematopoietic Stem Cell Transplantation ,Infant ,Histone-Lysine N-Methyltransferase ,Hematology ,Immunotherapy ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,medicine.disease ,Combined Modality Therapy ,Kidney Neoplasms ,Chimeric antigen receptor ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,KMT2A ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Pediatrics, Perinatology and Child Health ,biology.protein ,Cancer research ,Female ,Chimeric Antigen Receptor T-Cell Therapy ,Neoplasm Recurrence, Local ,Bone Marrow Neoplasms ,business ,Myeloid-Lymphoid Leukemia Protein ,030215 immunology - Abstract
Chimeric antigen receptor (CAR) T-cells, engineered autologous T-cells that target antigens found in leukemia, have shown durable remissions in relapsed acute lymphoblastic leukemia (ALL). Infant ALL with KMT2A rearrangements (KMT2Ar) is a rare, aggressive form of leukemia associated with extramedullary disease both at diagnosis and at relapse, and overall outcomes for these patients are dismal. Here we report the successful use of tisagenlecleucel, a CAR T-cell product approved for relapsed/refractory ALL, in a patient with KMT2Ar infant ALL who was treated for combined marrow and extramedullary (renal) relapse.
- Published
- 2020
- Full Text
- View/download PDF
18. High-Dose Methotrexate-Induced Idiopathic Intracranial Hypertension in Infant Acute Lymphoblastic Leukemia
- Author
-
Yazhi Zhang, Yining Qiu, Xiaoyan Wu, Louis Hinkle, Runming Jin, Zhujun Wang, and Ran Wang
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,Lymphoblastic Leukemia ,medicine.medical_treatment ,Case Report ,dexamethasone ,acute lymphoblastic leukemia ,Anterior fontanelle ,03 medical and health sciences ,0302 clinical medicine ,neurotoxicity ,medicine ,Pharmacology (medical) ,high-dose methotrexate ,skin and connective tissue diseases ,Dexamethasone ,Pharmacology ,Chemotherapy ,business.industry ,lcsh:RM1-950 ,Neurotoxicity ,medicine.disease ,High dose methotrexate ,Infant Acute Lymphoblastic Leukemia ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Methotrexate ,business ,idiopathic intracranial hypertension ,medicine.drug - Abstract
A 7-month-old baby girl with acute lymphoblastic leukemia (ALL) presented with bulging anterior fontanelle after completing the first and second courses of high-dose methotrexate (HD-MTX) chemotherapy. Between courses, the infant recovered and was discharged. Prior to the third and fourth HD-MTX courses, the baby girl was administered infusions of dexamethasone, which prevented recurrence of neurological side effects observed after the first and second courses of HD-MTX. To our knowledge, this is the first reported case of HD-MTX-induced idiopathic intracranial hypertension in infants, and that prophylactic use of dexamethasone can be applied to prevent acute intracranial hypertension following HD-MTX infusion.
- Published
- 2020
- Full Text
- View/download PDF
19. Clinical features and prognosis of infant acute lymphoblastic leukemia in China: A single-center retrospective analysis
- Author
-
Jianxin Li, Fang Tao, Yi Li, Zhi Chen, Hui Li, Kaili Li, Hao Xiong, Ping Zhou, and Zhuo Wang
- Subjects
Pediatrics ,medicine.medical_specialty ,Regimen ,business.industry ,medicine ,Retrospective analysis ,Risk factor ,Single Center ,business ,Childhood all ,Infant Acute Lymphoblastic Leukemia - Abstract
Background: In this retrospective analysis, we investigate the clinical features and prognosis of 23 infant patients ( 100 × 109/L at initial diagnosis as a risk factor for poor OS and EFS. Conclusion: Treatment of infant ALL with the standard childhood ALL regimen achieved an OS rate similar to patients with high-risk ALL, and WBC at initial diagnosis may be an important prognostic indicator.
- Published
- 2020
- Full Text
- View/download PDF
20. First report of t(5;11) KMT2A-MAML1 fusion in de novo infant acute lymphoblastic leukemia
- Author
-
Fabio Fuligni, Adam Shlien, James A. Whitlock, Nisha Kanwar, Scott Davidson, Anita Villani, Sneha Tandon, Mary Shago, and Oussama Abla
- Subjects
Oncology ,Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Poor prognosis ,Oncogene Proteins, Fusion ,Chromosomal translocation ,KMT2A Gene Rearrangement ,Newly diagnosed ,Translocation, Genetic ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Genetics ,medicine ,Humans ,Molecular Biology ,Gene ,Acute leukemia ,biology ,Chromosomes, Human, Pair 11 ,Infant ,Histone-Lysine N-Methyltransferase ,Prognosis ,Infant Acute Lymphoblastic Leukemia ,DNA-Binding Proteins ,KMT2A ,030220 oncology & carcinogenesis ,biology.protein ,Chromosomes, Human, Pair 5 ,Myeloid-Lymphoid Leukemia Protein ,Transcription Factors - Abstract
Infant acute lymphoblastic leukemia (ALL) comprises 2.5%-5% of pediatric ALL with inferior survival compared to older children. A majority of infants (80%) with ALL harbor KMT2A gene rearrangement, which portends a poor prognosis. Approximately 94 different partner genes have been identified to date. The common rearrangements include t(4;11)(q21;q23)KMT2A-AFF1,t(11;19) (q23;p13.3)KMT2A-MLLT1 and t(9;11)(p22;q23)KMT2A-MLLT3. We report a novel translocation t(5;11)(q35;q23)KMT2A-MAML1 in newly diagnosed infant precursor B-ALL. Long-term follow-up and a larger number of patients are needed to better understand its prognostic significance.
- Published
- 2020
21. Targeting EIF4E signaling with ribavirin in infant acute lymphoblastic leukemia
- Author
-
Carolyn A. Felix, Patrizia Porazzi, Joanne M. Hilden, James W Davenport, Sarah K. Tasian, Meenakshi Devidas, ZoAnn E. Dreyer, Shenghao Jin, I-Ming L. Chen, Alix E. Seif, Tiffaney Vincent, Andrew J. Carroll, Martin Carroll, Blaine W. Robinson, Karen A. Urtishak, David T. Teachey, Katherine L. B. Borden, Biljana Culjkovic-Kraljacic, Stephen P. Hunger, Li-San Wang, Jeffrey S. Barrett, Jonni S. Moore, Richard C. Harvey, Nyla A. Heerema, and Cheryl L. Willman
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,Indoles ,medicine.medical_treatment ,Biology ,Acute lymphoblastic leukemia ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,EIF4E ,Ribavirin ,Genetics ,medicine ,Humans ,MCL1 ,Pyrroles ,Molecular Targeted Therapy ,Infant, KMT2A ,Molecular Biology ,Chemotherapy ,Cell growth ,Gene Expression Regulation, Leukemic ,Gene Expression Profiling ,Infant ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Microarray Analysis ,3. Good health ,Infant Acute Lymphoblastic Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Eukaryotic Initiation Factor-4E ,chemistry ,Cell culture ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Child, Preschool ,Multigene Family ,Protein Biosynthesis ,Cancer research ,Bone marrow ,Obatoclax ,Signal Transduction - Abstract
The poor outcomes in infant acute lymphoblastic leukemia (ALL) necessitate new treatments. Here we discover that EIF4E protein is elevated in most cases of infant ALL and test EIF4E targeting by the repurposed antiviral agent ribavirin, which has anticancer properties through EIF4E inhibition, as a potential treatment. We find that ribavirin treatment of actively dividing infant ALL cells on bone marrow stromal cells (BMSCs) at clinically achievable concentrations causes robust proliferation inhibition in proportion with EIF4E expression. Further, we find that ribavirin treatment of KMT2A-rearranged (KMT2A-R) infant ALL cells and the KMT2A-AFF1 cell line RS4:11 inhibits EIF4E, leading to decreases in oncogenic EIF4E-regulated cell growth and survival proteins. In ribavirin-sensitive KMT2A-R infant ALL cells and RS4:11 cells, EIF4E-regulated proteins with reduced levels of expression following ribavirin treatment include MYC, MCL1, NBN, BCL2 and BIRC5. Ribavirin-treated RS4:11 cells exhibit impaired EIF4E-dependent nuclear to cytoplasmic export and/or translation of the corresponding mRNAs, as well as reduced phosphorylation of the p-AKT1, p-EIF4EBP1, p-RPS6 and p-EIF4E signaling proteins. This leads to an S-phase cell cycle arrest in RS4:11 cells corresponding to the decreased proliferation. Ribavirin causes nuclear EIF4E to re-localize to the cytoplasm in KMT2A-AFF1 infant ALL and RS4:11 cells, providing further evidence for EIF4E inhibition. Ribavirin slows increases in peripheral blasts in KMT2A-R infant ALL xenograft-bearing mice. Ribavirin cooperates with chemotherapy, particularly L-asparaginase, in reducing live KMT2A-AFF1 infant ALL cells in BMSC co-cultures. This work establishes that EIF4E is broadly elevated across infant ALL and that clinically relevant ribavirin exposures have preclinical activity and effectively inhibit EIF4E in KMT2A-R cases, suggesting promise in EIF4E targeting using ribavirin as a means of treatment.
- Published
- 2018
22. Trametinib inhibits RAS-mutant MLL-rearranged acute lymphoblastic leukemia at specific niche sites and reduces ERK phosphorylation in vivo
- Author
-
Priscilla Wander, Pauline Schneider, Rob Pieters, P. Castro, Mark Kerstjens, Ronald W. Stam, Sandra S. Pinhanços, and Pediatrics
- Subjects
0301 basic medicine ,Trametinib ,biology ,Mutant ,Hematology ,Gene rearrangement ,Infant Acute Lymphoblastic Leukemia ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,KMT2A ,Cell culture ,In vivo ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Gene - Abstract
The majority (approx. 80%) of infant acute lymphoblastic leukemia (ALL) cases are characterized by chromosomal rearrangements involving the Mixed Lineage Leukemia ( MLL , or KMT2A ) gene, which confer a poor prognosis: chances of event-free survival are 30–40% at best.[1][1] We and others showed
- Published
- 2018
23. Single-Cell Genomic Analysis Identifies Prognostically Significant Gene Expression Programs in Infant Acute Lymphoblastic Leukemia
- Author
-
Patrick A. Brown, Irina Pushel, Daniel Louiselle, Tomi Pastinen, Midhat S. Farooqi, Byunggil Yoo, and Erin M. Guest
- Subjects
medicine.anatomical_structure ,business.industry ,Immunology ,Gene expression ,Cell ,medicine ,Cancer research ,Cell Biology ,Hematology ,business ,Biochemistry ,Infant Acute Lymphoblastic Leukemia - Abstract
Introduction: Infant acute lymphoblastic leukemia (ALL) is an aggressive subtype of leukemia with low rates of survival. Rearrangements involving the KMT2A gene are associated with poor prognosis for infants with this cancer. Although many patients with KMT2A rearrangements (KMT2A-r) ultimately relapse, some do not. The molecular basis for this distinction has not yet been determined. There is evidence to suggest that cancers with distinct KMT2A fusion partners show distinct patterns of gene expression, though these differences have not previously been linked to prognostic outcomes. Here we utilize single-cell genomic analysis for infant ALL samples with and without KMT2A-r taken at diagnosis to explore transcriptional dynamics and identify gene expression programs with potential prognostic value. Methods: We performed 10x Chromium single-cell multiome sequencing to obtain both gene expression (scRNA-seq) and chromatin accessibility (scATAC-seq) data for blood and/or bone marrow samples obtained from a total of 34 infant ALL patients at time of diagnosis. Of these, 19 KMT2A-r patients later relapsed, 6 KMT2A-r patients did not relapse, and 9 patients did not show KMT2A rearrangement. Sequencing data were processed and aggregated with cellranger-arc, with normalization and differential gene expression performed using the Seurat package for gene expression analysis. Differential gene expression was performed between the three groups described above, (KMT2A-r + relapse, KMT2A-r no relapse, no KMT2A-r), then further subdivided for KMT2A-r cases based on the KMT2A partner gene (MLLT1 n=13 or AFF1 n=12). Results: In preliminary scRNA- and scATAC-seq in infant ALL patients, we had observed that cancer cells from individual patients tend to cluster separately, while non-blast cell populations showed similar interindividual patterns, suggesting a high degree of divergence among blast cell transcriptional programs. We replicated this finding here in larger patient samples, demonstrating no overt similarity based on KMT2A-r status or whether patients later relapsed. Strikingly, samples did broadly cluster according to KMT2A rearrangement partner gene, indicating that this appears to be a major driver of the transcriptional program in infant ALL patients. Differential expression analysis within the KMT2A-MLLT1 and KMT2A-AFF1 samples separately revealed expression of other genes associated with particular KMT2A fusion genes. Specifically, we observed that expression of HOXA genes was mostly restricted to KMT2A-MLLT1 samples. Consistent with previous results, we observed that within KMT2A-AFF1 samples there were two distinct groups of cells with mutually exclusive expression of HOXA9 or IRX1. Furthermore, the limited HOX gene expression observed in KMT2A-AFF1 samples was enriched in patients who did not relapse, supporting previous observations that the KMT2A-AFF1 samples expressing IRX1 comprise a more aggressive leukemia. Conclusions: Utilizing a single-cell transcriptomic approach enabled us to identify gene expression programs associated with good and poor prognosis in KMT2A-r infant ALL cases. We found that the KMT2A fusion partner gene appears to drive a large portion of the transcriptional heterogeneity observed across KMT2A-r samples. By treating the KMT2A-MLLT1 and KMT2A-AFF1 samples separately and exploring transcriptional dynamics within these groups, we identified transcriptional differences between patients who did and did not relapse in the presence of a KMT2A-AFF1 fusion. We are continuing to explore these data to identify prognostic markers in KMT2A-MLLT1 patients. Figure 1. Mutually exclusive gene expression programs distinguish subsets of infant ALL samples. A) Uniform manifold approximation and projection (UMAP) visualization of gene expression in individual cells across all patient samples at diagnosis, each color corresponding to a single patient (n=34). B) Cells colored according to KMT2A-r status and relapse or lack thereof. C) Cells colored according to KMT2A fusion partner gene. D) HOXA9 expression among KMT2A-AFF1 patients is strongly biased to patients that do not relapse. E) In KMT2A-AFF1 samples, HOXA9 and IRX1 show mutually exclusive expression patterns. Figure 1 Figure 1. Disclosures Brown: Novartis: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees; Kura: Membership on an entity's Board of Directors or advisory committees; KIte: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2021
- Full Text
- View/download PDF
24. Outcome of risk-based therapy for infant acute lymphoblastic leukemia with or without an MLL gene rearrangement, with emphasis on late effects: a final report of two consecutive studies, MLL96 and MLL98, of the Japan Infant Leukemia Study Group.
- Author
-
Tomizawa, D., Koh, K., Sato, T., Kinukawa, N., Morimoto, A., Isoyama, K., Kosaka, Y., Oda, T., Oda, M., Hayashi, Y., Eguchi, M., Horibe, K., Nakahata, T., Mizutani, S., and Ishii, E.
- Subjects
- *
LYMPHOBLASTIC leukemia in children , *LYMPHOBLASTIC leukemia , *STEM cell transplantation , *THERAPEUTICS , *DRUG therapy , *LEUKEMIA treatment - Abstract
We evaluated the efficacy of a treatment strategy in which infants with acute lymphoblastic leukemia (ALL) were stratified by their MLL gene status and then assigned to different risk-based therapies. A total of 102 patients were registered on two consecutive multicenter trials, designated MLL96 and MLL98, between 1995 and 2001. Those with a rearranged MLL gene (MLL-R, n=80) were assigned to receive intensive chemotherapy followed by hematopoietic stem cell transplantation (HSCT), while those with germline MLL (MLL-G, n=22) were treated with chemotherapy alone. The 5-year event-free survival (EFS) rate for all 102 infants was 50.9% (95% confidence interval, 41.0–60.8%). The most prominent late effect was growth impairment, observed in 58.9% of all evaluable patients in the MLL-R group. This plan of risk-based therapy appears to have improved the overall prognosis for infants with ALL, compared with previously reported results. However, over half the events in patients with MLL rearrangement occurred before the instigation of HSCT, and that HSCT-related toxic events comprised 36.3% (8/22) of post-transplantation events, suggesting that further stratification within the MLL-R group and the development of more effective early-phase intensification chemotherapy will be needed before the full potential of this strategy is realized.Leukemia (2007) 21, 2258–2263; doi:10.1038/sj.leu.2404903; published online 9 August 2007 [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
25. Death Within 1 Month of Diagnosis in Childhood Cancer: An Analysis of Risk Factors and Scope of the Problem
- Author
-
Karina Braga Ribeiro, Adam L. Green, Elissa Furutani, and Carlos Rodriguez Galindo
- Subjects
0301 basic medicine ,Cancer Research ,Pediatrics ,medicine.medical_specialty ,Adolescent ,Population ,Disease ,Cohort Studies ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Neoplasms ,medicine ,Humans ,Young adult ,Child ,education ,Retrospective Studies ,education.field_of_study ,business.industry ,Infant ,Cancer ,Retrospective cohort study ,ORIGINAL REPORTS ,Odds ratio ,medicine.disease ,United States ,Infant Acute Lymphoblastic Leukemia ,030104 developmental biology ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,business ,SEER Program ,Cohort study - Abstract
Purpose Despite advances in childhood cancer care, some patients die soon after diagnosis. This population is not well described and may be under-reported. Better understanding of risk factors for early death and scope of the problem could lead to prevention of these occurrences and thus better survival rates in childhood cancer. Methods We retrieved data from SEER 13 registries on 36,337 patients age 0 to 19 years diagnosed with cancer between 1992 and 2011. Early death was defined as death within 1 month of diagnosis. Socioeconomic status data for each individual’s county of residence were derived from Census 2000. Crude and adjusted odds ratios and corresponding 95% CIs were estimated for the association between early death and demographic, clinical, and socioeconomic factors. Results Percentage of early death in the period was 1.5% (n = 555). Children with acute myeloid leukemia, infant acute lymphoblastic leukemia, hepatoblastoma, and malignant brain tumors had the highest risk of early death. On multivariable analysis, an age younger than 1 year was a strong predictor of early death in all disease groups examined. Black race and Hispanic ethnicity were both risk factors for early death in multiple disease groups. Residence in counties with lower than median average income was associated with a higher risk of early death in hematologic malignancies. Percentages of early death decreased significantly over time, especially in hematologic malignancies. Conclusion Risk factors for early death in childhood cancer include an age younger than 1 year, specific diagnoses, minority race and ethnicity, and disadvantaged socioeconomic status. The population-based disease-specific percentages of early death were uniformly higher than those reported in cooperative clinical trials, suggesting that early death is under-reported in the medical literature. Initiatives to identify those at risk and develop preventive interventions should be prioritized.
- Published
- 2017
- Full Text
- View/download PDF
26. MEK inhibition is a promising therapeutic strategy for MLL-rearranged infant acute lymphoblastic leukemia patients carrying RAS mutations
- Author
-
Sandra S. Pinhanços, Merel Willekes, Pauline Schneider, Mark Kerstjens, Rob Pieters, Emma M. C. Driessen, Ronald W. Stam, and Pediatrics
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Mutant ,MAP Kinase Kinase 1 ,Apoptosis ,RAS-pathway ,Translocation, Genetic ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Tumor Cells, Cultured ,Humans ,MEK inhibitors ,Protein Kinase Inhibitors ,Cell Proliferation ,Trametinib ,Gene Rearrangement ,Hematology ,business.industry ,Wild type ,leukemia ,Infant ,Histone-Lysine N-Methyltransferase ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,MLL-rearrangements ,030104 developmental biology ,Oncology ,Ras Signaling Pathway ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Immunology ,Mutation ,Selumetinib ,Cancer research ,ras Proteins ,business ,Myeloid-Lymphoid Leukemia Protein ,Research Paper - Abstract
// Mark Kerstjens 1, * , Emma M.C. Driessen 1, * , Merel Willekes 1 , Sandra S. Pinhancos 1 , Pauline Schneider 1 , Rob Pieters 1, 2 , Ronald W. Stam 1 1 Department of Pediatric Oncology/Hematology, Erasmus MC-Sophia Children’s Hospital, Rotterdam, The Netherlands 2 Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands * These authors have contributed equally to this work Correspondence to: Ronald W. Stam, email: r.stam@erasmusmc.nl Keywords: MLL-rearrangements, RAS-pathway, leukemia, MEK inhibitors Received: May 31, 2016 Accepted: August 13, 2016 Published: August 31, 2016 ABSTRACT Acute lymphoblastic leukemia (ALL) in infants is an aggressive malignancy with a poor clinical outcome, and is characterized by translocations of the Mixed Lineage Leukemia (MLL) gene. Previously, we identified RAS mutations in 14-24% of infant ALL patients, and showed that the presence of a RAS mutation decreased the survival chances even further. We hypothesized that targeting the RAS signaling pathway could be a therapeutic strategy for RAS -mutant infant ALL patients. Here we show that the MEK inhibitors Trametinib, Selumetinib and MEK162 severely impair primary RAS -mutant MLL -rearranged infant ALL cells in vitro . While all RAS -mutant samples were sensitive to MEK inhibitors, we found both sensitive and resistant samples among RAS -wildtype cases. We confirmed enhanced RAS pathway signaling in RAS -mutant samples, but found no apparent downstream over-activation in the wildtype samples. However, we did confirm that MEK inhibitors reduced p-ERK levels, and induced apoptosis in the RAS -mutant MLL -rearranged ALL cells. Finally, we show that MEK inhibition synergistically enhances prednisolone sensitivity, both in RAS -mutant and RAS -wildtype cells. In conclusion, MEK inhibition represents a promising therapeutic strategy for MLL -rearranged ALL patients harboring RAS mutations, while patients without RAS mutations may benefit through prednisolone sensitization.
- Published
- 2017
27. No benefit of Interfant protocols compared to BFM-based protocols for infants with acute lymphoblastic leukemia. Results from an institution in Argentina
- Author
-
Jorge Rossi, Edgardo M Baialardo, Patricia L. Rubio, María A. Deu, Cristian G. Sánchez La Rosa, Elizabeth M. Alfaro, Pedro Zubizarreta, Carla L Pennella, Maria S. Felice, Myriam Guitter, and Cristina N. Alonso
- Subjects
Male ,medicine.medical_specialty ,medicine.medical_treatment ,Disease ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Retrospective Studies ,Protocol (science) ,Chemotherapy ,business.industry ,Infant ,Retrospective cohort study ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,Minimal residual disease ,Infant Acute Lymphoblastic Leukemia ,Survival Rate ,Standard error ,Oncology ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Female ,Outcomes research ,Neoplasm Recurrence, Local ,business ,030215 immunology ,Follow-Up Studies - Abstract
Background Infant acute lymphoblastic leukemia (ALL) is an infrequent disease characterized by clinical and biological features related to poor prognosis. Adapted therapies were designed without a clear consensus regarding the best treatment options. We aimed to compare the outcome between infant ALL cases receiving Interfant versus BFM-based protocols. Procedure This is a retrospective observational study. From April 1990 to June 2018, infant ALL cases were enrolled in one of the five consecutive treatment protocols. Clinical, demographic, and biological features and outcome were evaluated. A comparative analysis was performed between Interfant protocols and BFM-based protocols. Results During the studied period, 1913 ALL patients were admitted and 116 (6%) were infants. Treatment administered was: ALL-BFM'90 (n = 16), 1-ALL96-BFM/HPG (n = 7), Interfant-99 (n = 39), Interfant-06 (n = 35), and ALLIC-BFM'2009 (n = 19). The 5-year event-free survival probability (EFSp) was 31.9(standard error [SE] 4.6)% for the entire population, with a significant difference among risk groups according to Interfant-06 criteria (P = .0029). KMT2A-rearrangement status was the strongest prognostic factor (P = .048), independently of the protocol strategy. The median time for relapse was 24.1 months for patients with minimal residual disease (MRD)-negative versus 11.5 months for those with MRD-positive (P = .0386). EFSp and cumulative relapse risk probability (CRRp) were similar. Interfant protocols showed comparable induction (8.1% vs 7.1%, P = .852) and complete remission mortality (21.6% vs 28.6%, P = .438), failing to reduce the relapse rate (48.5% vs 30.7%, P = .149). Conclusions Interfant protocols and BFM-based protocols presented comparable results. The risk group stratification proposed by Interfant-06 was validated by our results, and MRD seems useful to identify patients with an increased risk of early relapse.
- Published
- 2019
28. Peer Review #1 of 'Identification of hub genes and molecular mechanisms in infant acute lymphoblastic leukemia with MLL gene rearrangement (v0.2)'
- Author
-
D Neelakantan
- Subjects
Hub genes ,Genetics ,Identification (biology) ,Biology ,Infant Acute Lymphoblastic Leukemia ,Mll gene - Published
- 2019
- Full Text
- View/download PDF
29. Differential network analysis and protein-protein interaction study reveals active protein modules in glucocorticoid resistance for infant acute lymphoblastic leukemia
- Author
-
Abbas Nowzari-Dalini, Ali Masoudi-Nejad, Yasir Rahmatallah, and Zaynab Mousavian
- Subjects
0301 basic medicine ,Systems biology ,Context (language use) ,Computational biology ,Acute lymphoblastic leukemia ,Protein–protein interaction ,lcsh:Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Genetics ,Humans ,Gene Regulatory Networks ,lcsh:QD415-436 ,Databases, Protein ,Glucocorticoids ,Molecular Biology ,Gene ,Genetics (clinical) ,biology ,Gene Expression Profiling ,lcsh:RM1-950 ,Differential co-expression network analysis ,Histone-Lysine N-Methyltransferase ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Phenotype ,Infant Acute Lymphoblastic Leukemia ,Active protein modules ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,KMT2A ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,biology.protein ,Glucocorticoid resistance ,Molecular Medicine ,Myeloid-Lymphoid Leukemia Protein ,Research Article ,Signal Transduction - Abstract
Background Acute lymphoblastic leukemia (ALL) is the most common type of cancer diagnosed in children and Glucocorticoids (GCs) form an essential component of the standard chemotherapy in most treatment regimens. The category of infant ALL patients carrying a translocation involving the mixed lineage leukemia (MLL) gene (gene KMT2A) is characterized by resistance to GCs and poor clinical outcome. Although some studies examined GC-resistance in infant ALL patients, the understanding of this phenomenon remains limited and impede the efforts to improve prognosis. Methods This study integrates differential co-expression (DC) and protein-protein interaction (PPI) networks to find active protein modules associated with GC-resistance in MLL-rearranged infant ALL patients. A network was constructed by linking differentially co-expressed gene pairs between GC-resistance and GC-sensitive samples and later integrated with PPI networks by keeping the links that are also present in the PPI network. The resulting network was decomposed into two sub-networks, specific to each phenotype. Finally, both sub-networks were clustered into modules using weighted gene co-expression network analysis (WGCNA) and further analyzed with functional enrichment analysis. Results Through the integration of DC analysis and PPI network, four protein modules were found active under the GC-resistance phenotype but not under the GC-sensitive. Functional enrichment analysis revealed that these modules are related to proteasome, electron transport chain, tRNA-aminoacyl biosynthesis, and peroxisome signaling pathways. These findings are in accordance with previous findings related to GC-resistance in other hematological malignancies such as pediatric ALL. Conclusions Differential co-expression analysis is a promising approach to incorporate the dynamic context of gene expression profiles into the well-documented protein interaction networks. The approach allows the detection of relevant protein modules that are highly enriched with DC gene pairs. Functional enrichment analysis of detected protein modules generates new biological hypotheses and may help in explaining the GC-resistance in MLL-rearranged infant ALL patients. Electronic supplementary material The online version of this article (10.1186/s10020-019-0106-1) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
30. Cryptic recurrent ACIN1-NUTM1 fusions in non-KMT2A-rearranged infant acute lymphoblastic leukemia
- Author
-
Josette-Renée Landry, Louise Laramée, Brian T. Wilhelm, Josée Hébert, Loubna Jouan, Françoise Couture, Alexandre Rouette, Henrique Bittencourt, Sonia Cellot, Luc L. Oligny, Thomas Pincez, Mélanie Bilodeau, Daniel Sinnett, Mathieu Roussy, Thai Hoa Tran, and Patrick Gendron
- Subjects
Male ,Cancer Research ,Oncogene Proteins, Fusion ,Fusion gene ,03 medical and health sciences ,Exon ,Cytogenetics ,0302 clinical medicine ,Genetics ,medicine ,NUTM1 Gene ,Humans ,Gene ,Chromosome Aberrations ,Gene Rearrangement ,biology ,Infant, Newborn ,Nuclear Proteins ,Histone-Lysine N-Methyltransferase ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Minimal residual disease ,Immunohistochemistry ,Infant Acute Lymphoblastic Leukemia ,Neoplasm Proteins ,Leukemia ,Leukemia, Myeloid, Acute ,KMT2A ,030220 oncology & carcinogenesis ,biology.protein ,Gene Fusion ,Myeloid-Lymphoid Leukemia Protein - Abstract
Infant acute lymphoblastic leukemias (ALL) are rare hematological malignancies occurring in children younger than 1 year of age, most frequently associated with KMT2A rearrangements (KMT2A-r). The smaller subset without KMT2A-r, which represents 20% of infant ALL cases, is poorly characterized. Here we report two cases of chemotherapy-sensitive non-KMT2A-r infant ALL. Transcriptome analyses revealed identical ACIN1-NUTM1 gene fusions in both cases, derived from cryptic chromosomal rearrangements undetected by standard cytogenetic approaches. Two isoforms of the gene fusion, joining exons 3 or 4 of ACIN1 to exon 3 of NUTM1, were identified. Both fusion transcripts contained the functional DNA-binding SAP (SAF-A/B, Acinus, and PIAS) domain of ACIN1 and most of NUTM1. The detection of the ACIN1-NUTM1 fusion by RT-PCR allowed the molecular monitoring of minimal residual disease in a clinical setting. Based on publicly available genomic datasets and literature review, we predict that NUTM1 gene fusions are recurrent events in infant ALL. As such, we propose two clinically relevant assays to screen for NUTM1 rearrangements in bone marrow cells, independent of the fusion partner: NUMT1 immunohistochemistry and NUTM1 RNA expression. In sum, our study identifies ACIN1-NUTM1 as a recurrent and possibly cryptic fusion in non-KMT2A-r infant ALL, provides clinical tools to screen for NUTM1-rearranged leukemia and contributes to the refinement of this new subgroup.
- Published
- 2019
31. Whole transcriptome sequencing reveals a KMT2A-USP2 fusion in infant acute myeloid leukemia
- Author
-
Shinichi Tsujimoto, Kohji Okamura, Junji Ikeda, Kazuhiko Nakabayashi, Shuichi Ito, Nobutaka Kiyokawa, Norio Shiba, Motohiro Kato, Kenichiro Hata, Hiroko Ogata-Kawata, Masanori Yoshida, Masanobu Takeuchi, Tomoo Osumi, and Daisuke Tomizawa
- Subjects
Male ,Cancer Research ,Poor prognosis ,Methyltransferase ,Whole Transcriptome Sequencing ,Chromosomal translocation ,Bioinformatics ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Endopeptidases ,Genetics ,Humans ,Oncogene Fusion ,biology ,Pediatric acute myeloid leukemia ,Myeloid leukemia ,Infant ,Histone-Lysine N-Methyltransferase ,Infant Acute Lymphoblastic Leukemia ,Leukemia, Myeloid, Acute ,KMT2A ,030220 oncology & carcinogenesis ,biology.protein ,Transcriptome ,Ubiquitin Thiolesterase ,Myeloid-Lymphoid Leukemia Protein - Abstract
Infant acute lymphoblastic leukemia with lysine (K)-specific methyltransferase 2A (KMT2A) rearrangements usually has a poor prognosis regardless of the fusion partners of KMT2A. However, the prognosis of pediatric acute myeloid leukemia (AML) with KMT2A rearrangements depends on its translocation partners. We herein report the case of a 9-month-old boy with a KMT2A-USP2 fusion, which required diagnosis by whole transcriptome sequencing after the failure of detection of known translocation partners by conventional screening approaches. As this first report of a patient with AML with a KMT2A-USP2 fusion illustrates, identification of the partners in all patients with KMT2A-rearranged AML is critical to elucidate the outcomes associated with specific rearrangements and to develop appropriate treatment strategies. Moreover, development of additional methods to detect specific translocation partners of KMT2A and leukemia-specific targeting drugs is important to improve further the outcomes of KMT2A-rearranged AML.
- Published
- 2019
32. Infant lymphoblastic Leukemia: A Single Centers 10 Year Experience
- Author
-
Fatma Gumruk, Mualla Cetin, Selin Aytac, Sule Unal, Duygu Uckan-Cetinkaya, İnci Yaman-Bajin, Baris Kuskonmaz, and Çocuk Sağlığı ve Hastalıkları
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,Turkey ,medicine.medical_treatment ,Lymphoblastic Leukemia ,Improved survival ,Hematopoietic stem cell transplantation ,Hospitals, University ,Recurrence ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Childhood all ,Retrospective Studies ,Chemotherapy ,Triplets ,business.industry ,Remission Induction ,Hematopoietic Stem Cell Transplantation ,Infant, Newborn ,Infant ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Infant Acute Lymphoblastic Leukemia ,Transplantation ,Pediatrics, Perinatology and Child Health ,Female ,Infant leukemia ,business ,Rare disease - Abstract
Yaman-Bajin İ, Aytaç S, Kuşkonmaz B, Uçkan-Çetinkaya D, Ünal Ş, Gümrük F, Çetin M. Infant lymphoblastic leukemia: a single centers 10 year experience. Turk J Pediatr 2019; 61: 325-329. Infant acute lymphoblastic leukemia (ALL) is a rare disease and consists of 4-5% of all childhood ALL. Despite improved survival rates in childhood ALL, infants with ALL have a worse prognosis. We aimed to evaluate the clinical features and treatment outcomes of our patients diagnosed with infant ALL at Hacettepe University, Pediatric Hematology Department between 1 January 2008 and 1 January 2018 retrospectively. There were 13 patients with a median age of 7 months. Three of the patients were triplets born from a spontaneous monozygotic triplet pregnancy. Relapse were observed in 4 patients. Hematopoietic stem cell transplantation (HSCT) was performed for five patients. Relapse after HSCT was observed in 3 patients. After a median follow-up period of 18 months, 6 patients (45%) (3 after HSCT and 3 who only received chemotherapy) were alive and in remission. Prognosis of infant ALL is poor in that only half of the patients survive. Our results suggest that bone marrow transplantation seems to be a good and efficient choice of treatment for selected patients. However, there is still a big issue to decide which patient should undergo transplantation and more studies are needed to reevaluate the eligibility criteria for HSCT in this group of patients.
- Published
- 2019
33. Resolving driver events in MLL-r negative high-risk infant ALL
- Author
-
Ledia Brunga, Fabio Fuligni, Jennifer Seelisch, Adam Shlien, Sumit Gupta, Federico Comitani, Matthew Zatzman, and Patrick A. Brown
- Subjects
Cancer Research ,Pediatrics ,medicine.medical_specialty ,Oncology ,business.industry ,hemic and lymphatic diseases ,Medicine ,business ,neoplasms ,Childhood all ,High risk infants ,Infant Acute Lymphoblastic Leukemia - Abstract
10030 Background: Infant acute lymphoblastic leukemia (ALL) is the only subtype of childhood ALL whose outcome has not improved over the past two decades. The most important prognosticator is the presence of rearrangements in the Mixed Lineage Leukemia gene (MLL-r), however, many patients present with high-risk clinical features but without MLL-r. We recently identified two cases of infant ALL with high-risk clinical features resembling MLL-r, but were negative for MLL-r by conventional diagnostics. RNA sequencing revealed a partial tandem duplication in MLL (MLL-PTD). We thus aimed to determine if MLL-PTD, other MLL abnormalities, or other genetic or transcriptomic features were driving this subset of high-risk infant ALL without MLL-r. Methods: We obtained 19 banked patient samples from the Children’s Oncology Group (COG) infant ALL trial (AALL0631) from MLL wildtype patients as determined by FISH and cytogenetics. Utilizing deep RNA-sequencing, we manually inspected the MLL gene for MLL-PTD, while also performing automated fusion detection and gene expression profiling in search of defining features of these tumors. Results: 3 additional MLL-PTDs were identified, all in patients with infant T-cell ALL, whereas both index cases were in patients with infant B-cell ALL. Gene expression profiling analysis revealed that all five MLL-PTD infants clustered together. Eight infants (7 with B-cell ALL) were found to have Ph-like expression. Five of these 8 infants were also found to have an IKZF1/JAK2 expression profile; one of these five had a PAX5-JAK2 fusion detected. Two infants (including the one noted above) had novel PAX5 fusions, known drivers of B-cell leukemia. Additional detected fusions included TCF3-PBX1 and TCF4-ZNF384. Conclusions: MLL-PTDs were found in both B- and T-cell infant ALL. Though Ph-like ALL has been described in adolescents and young adults, we found a substantial frequency of Ph-like expression among MLL-WT infants. Further characterization of these infants is ongoing. If replicated in other infant cohorts, these two findings may help explain the poor prognosis of MLL-WT ALL when compared to children with standard risk ALL, and offer the possibility of targeted therapy for select infants.
- Published
- 2021
- Full Text
- View/download PDF
34. Correction to: FLT3 inhibitor lestaurtinib plus chemotherapy for newly diagnosed KMT2A-rearranged infant acute lymphoblastic leukemia: Children’s Oncology Group trial AALL0631
- Author
-
Mignon L. Loh, Naomi J. Winick, Nyla A. Heerema, Andrew J. Carroll, Lia Gore, Zo Ann E. Dreyer, John A. Kairalla, Cindy Wang, Elizabeth A. Raetz, William L. Carroll, Michael J. Borowitz, Stephen P. Hunger, Patrick A. Brown, Joanne M. Hilden, Meenakshi Devidas, Donald Small, and Wanda L. Salzer
- Subjects
Oncology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,Group trial ,biology ,business.industry ,Lestaurtinib ,medicine.medical_treatment ,Hematology ,Newly diagnosed ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,KMT2A ,Internal medicine ,biology.protein ,Medicine ,business ,FLT3 Inhibitor ,medicine.drug - Published
- 2021
- Full Text
- View/download PDF
35. HOXA9/IRX1 expression pattern defines two subgroups of infant MLL-AF4-driven acute lymphoblastic leukemia
- Author
-
Katrin Ottersbach and Vasiliki Symeonidou
- Subjects
0301 basic medicine ,IRX1 ,Cancer Research ,Adolescent ,Oncogene Proteins, Fusion ,Cell of origin ,Disease ,Biology ,Brief Communication ,Bioinformatics ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Child ,Molecular Biology ,Gene ,Homeodomain Proteins ,Gene Expression Regulation, Leukemic ,Infant ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,030104 developmental biology ,030220 oncology & carcinogenesis ,Homeobox ,Myeloid-Lymphoid Leukemia Protein ,Transcription Factors - Abstract
Highlights • We identified two sub-groups of infant MLL-AF4-driven ALL, iALL-HOXA9, and iALL-IRX1. • The subgroups exhibit mutually exclusive expression of HOXA9/HOXA10 and IRX1. • The transcriptional profile of iALL-IRX1 patients revealed a more aggressive disease. • The two subgroups exhibit different expression of potential therapeutic targets., Infant t(4;11) acute lymphoblastic leukemia is the most common leukemia in infant patients and has a highly aggressive nature. The patients have a dismal prognosis, which has not improved in more than a decade, suggesting that a better understanding of this disease is required. In the study described here, we analyzed two previously published RNA-sequencing data sets and gained further insights into the global transcriptomes of two known subgroups of this disease, which are characterized by the presence or absence of a homeobox gene expression signature. Specifically, we identified a remarkable mutually exclusive expression of the HOXA9/HOXA10 and IRX1 genes and termed the two subgroups iALL-HOXA9 and iALL-IRX1. This expression pattern is critical as it suggests that there is a fundamental difference between the two subgroups. Investigation of the transcriptomes of the two subgroups reveals a more aggressive nature for the iALL-IRX1 group, which is further supported by the fact that patients within this group have a worse prognosis and are also diagnosed at a younger age. This could be reflective of a developmentally earlier cell of origin for iALL-IRX1. Our analysis further uncovered critical differences between the two groups that may have an impact on treatment strategies. In summary, after a detailed investigation into the transcriptional profiles of iALL-HOXA9 and iALL-IRX1 patients, we highlight the importance of acknowledging that these two subgroups are different and that this is of clinical importance.
- Published
- 2021
- Full Text
- View/download PDF
36. Network-based expression analysis reveals key genes related to glucocorticoid resistance in infant acute lymphoblastic leukemia
- Author
-
Ali Masoudi-Nejad, Abbas Nowzari-Dalini, Zaynab Mousavian, Ronald W Stam, and Yasir Rahmatallah
- Subjects
0301 basic medicine ,Cancer Research ,Candidate gene ,Gene regulatory network ,Antineoplastic Agents ,Biology ,03 medical and health sciences ,microRNA ,Humans ,Gene Regulatory Networks ,Glucocorticoids ,Gene ,Genetics ,Gene Expression Profiling ,Infant ,Histone-Lysine N-Methyltransferase ,General Medicine ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Infant Acute Lymphoblastic Leukemia ,Gene expression profiling ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Molecular Medicine ,Myeloid-Lymphoid Leukemia Protein ,Gene co-expression network - Abstract
Despite vast improvements that have been made in the treatment of children with acute lymphoblastic leukemia (ALL), the majority of infant ALL patients (~80 %, < 1 year of age) that carry a chromosomal translocation involving the mixed lineage leukemia (MLL) gene shows a poor response to chemotherapeutic drugs, especially glucocorticoids (GCs), which are essential components of all current treatment regimens. Although addressed in several studies, the mechanism(s) underlying this phenomenon have remained largely unknown. A major drawback of most previous studies is their primary focus on individual genes, thereby neglecting the putative significance of inter-gene correlations. Here, we aimed at studying GC resistance in MLL-rearranged infant ALL patients by inferring an associated module of genes using co-expression network analysis. The implications of newly identified candidate genes with associations to other well-known relevant genes from the same module, or with associations to known transcription factor or microRNA interactions, were substantiated using literature data. A weighted gene co-expression network was constructed to identify gene modules associated with GC resistance in MLL-rearranged infant ALL patients. Significant gene ontology (GO) terms and signaling pathways enriched in relevant modules were used to provide guidance towards which module(s) consisted of promising candidates suitable for further analysis. Through gene co-expression network analysis a novel set of genes (module) related to GC-resistance was identified. The presence in this module of the S100 and ANXA genes, both well-known biomarkers for GC resistance in MLL-rearranged infant ALL, supports its validity. Subsequent gene set net correlation analyses of the novel module provided further support for its validity by showing that the S100 and ANXA genes act as ‘hub’ genes with potentially major regulatory roles in GC sensitivity, but having lost this role in the GC resistant phenotype. The detected module implicates new genes as being candidates for further analysis through associations with known GC resistance-related genes. From our data we conclude that available systems biology approaches can be employed to detect new candidate genes that may provide further insights into drug resistance of MLL-rearranged infant ALL cases. Such approaches complement conventional gene-wise approaches by taking putative functional interactions between genes into account.
- Published
- 2016
- Full Text
- View/download PDF
37. Systematic chemical and molecular profiling of MLL-rearranged infant acute lymphoblastic leukemia reveals efficacy of romidepsin
- Author
-
John O’Reilly, Kim W. Carter, Sajla Singh, Denise Anderson, Julia E. Wells, Timothy W. Failes, Mark N. Cruickshank, Ursula R. Kees, Timo Lassmann, N Smithers, Jette Ford, Laurence C. Cheung, Julian Ik-Tsen Heng, Catherine H. Cole, Rishi S. Kotecha, Alexander M. Gout, Rab K. Prinjha, and Greg M. Arndt
- Subjects
0301 basic medicine ,Cancer Research ,Romidepsin ,03 medical and health sciences ,Mice ,Cell Line, Tumor ,Depsipeptides ,Medicine ,Animals ,Humans ,Gene Rearrangement ,Antibiotics, Antineoplastic ,business.industry ,Infant, Newborn ,Hematology ,Gene rearrangement ,Cytidine deaminase ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Cyclin-Dependent Kinases ,Infant Acute Lymphoblastic Leukemia ,Histone Deacetylase Inhibitors ,Leukemia ,030104 developmental biology ,Oncology ,Immunology ,Cancer research ,Cytarabine ,Myeloid-Lymphoid Leukemia Protein ,Heterografts ,Original Article ,Histone deacetylase ,business ,Proteasome Inhibitors ,medicine.drug - Abstract
To address the poor prognosis of mixed lineage leukemia (MLL)-rearranged infant acute lymphoblastic leukemia (iALL), we generated a panel of cell lines from primary patient samples and investigated cytotoxic responses to contemporary and novel Food and Drug Administration-approved chemotherapeutics. To characterize representation of primary disease within cell lines, molecular features were compared using RNA-sequencing and cytogenetics. High-throughput screening revealed variable efficacy of currently used drugs, however identified consistent efficacy of three novel drug classes: proteasome inhibitors, histone deacetylase inhibitors and cyclin-dependent kinase inhibitors. Gene expression of drug targets was highly reproducible comparing iALL cell lines to matched primary specimens. Histone deacetylase inhibitors, including romidepsin (ROM), enhanced the activity of a key component of iALL therapy, cytarabine (ARAC) in vitro and combined administration of ROM and ARAC to xenografted mice further reduced leukemia burden. Molecular studies showed that ROM reduces expression of cytidine deaminase, an enzyme involved in ARAC deactivation, and enhances the DNA damage-response to ARAC. In conclusion, we present a valuable resource for drug discovery, including the first systematic analysis of transcriptome reproducibility in vitro, and have identified ROM as a promising therapeutic for MLL-rearranged iALL.
- Published
- 2016
38. Unique FamilialMLL(KMT2A)-Rearranged Precursor B-Cell Infant Acute Lymphoblastic Leukemia in Non-twin Siblings
- Author
-
Li-San Wang, Margaret D. Sarezky, Jaclyn A. Biegel, Karen A. Urtishak, Donna Wilmoth, Julie W. Stern, Eric F. Rappaport, Carolyn A. Felix, Blaine W. Robinson, and Kim E. Nichols
- Subjects
0301 basic medicine ,Genetics ,Proband ,Chromosomal translocation ,Karyotype ,Hematology ,Gene rearrangement ,Biology ,Zygosity ,Infant Acute Lymphoblastic Leukemia ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,KMT2A ,Oncology ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Immunology ,Complex Karyotype ,biology.protein - Abstract
Background Infant acute lymphoblastic leukemia (ALL) has never occurred in families except for the ∼100% concordant cases in monozygous twins attributed to twin-to-twin metastases. We report the first kindred with infant ALL in non-twin siblings. The siblings were diagnosed with MLL-rearranged (MLL-R) ALL 26 months apart. The second affected sibling had an unaffected dichorionic monozygous co-twin. Both had fatal outcomes. Procedures Translocations were characterized by karyotype, FISH, multiplex FISH, and MLL breakpoint cluster region (bcr) Southern blot analysis. Breakpoint junctions and fusion transcripts were cloned by PCR. TP53 mutation and NADPH quinone oxidorecuctase 1 (NQO1) C609T analyses were performed, and pedigree history and parental occupations were ascertained. The likelihood of chance occurrence of infant ALL in non-twin siblings was computed based on a binomial distribution. Zygosity was determined by single nucleotide polymorphism (SNP) array. Results The translocations were not related or vertically transmitted. The complex karyotype of the proband's ALL had chromosome 2, 3, 4, and 11 abnormalities causing a 5′-MLL-AFF1-3′ fusion and a non-productive rearrangement of 3′MLL with a chromosome 3q intergenic region. The affected twin's ALL exhibited a simple t(4;11). The complex karyotype of the proband's ALL suggested a genotoxic insult, but no exposure was identified. There was no germline TP53 mutation. The NQO1 C609T risk allele was absent. The likelihood of infant ALL occurring in non-twin siblings by chance alone is one in 1.198 × 109 families. Conclusions Whether because of a deleterious transplacental exposure, novel predisposition syndrome, or exceedingly rare chance occurrence, MLL-R infant ALL can occur in non-twin siblings. The discordant occurrence of infant ALL in the monozygous twins was likely because they were dichorionic.
- Published
- 2016
- Full Text
- View/download PDF
39. Germline Variants Associated with Cancer Predisposition and Bone Marrow Failure Are Common in KMT2A-r Infant Acute Lymphoblastic Leukemia Patients
- Author
-
Azhar Saeed, Emily G. Farrow, Byunggil Yoo, Sarah Mc Dermott, Erin M. Guest, Midhat S. Farooqi, Patrick A. Brown, and Neil A. Miller
- Subjects
Oncology ,medicine.medical_specialty ,biology ,business.industry ,Cancer predisposition ,Immunology ,Bone marrow failure ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Germline ,Infant Acute Lymphoblastic Leukemia ,KMT2A ,Internal medicine ,biology.protein ,Medicine ,business - Abstract
Introduction: Infant acute lymphoblastic leukemia (ALL), is a particularly aggressive subtype of leukemia with an early onset and unfavorable clinical outcome. Most (~70%) cases of infant ALL involve chromosomal rearrangement of KMT2A (KMT2A-r) on chromosome 11q23, the strongest independent predictor of a poor prognosis. To date, genomics studies have consistently demonstrated KMT2A-r infant ALL to have a strikingly silent landscape of DNA mutations, aside from the KMT2A-r itself. Germline mutations in cancer predisposition genes are found in 8.6% of pediatric malignancies and 4.4% of pediatric leukemias, compared to 1.1% in persons in the 1000 Genomes Project (Zhang J et al., N Engl J Med 2015). We hypothesized that germline variants may contribute to the development of KMT2A-r ALL in infants. We examined the germline variants in remission blood samples from a large cohort of infants with KMT2A-r ALL who were enrolled in Children's Oncology Group (COG) trial AALL15P1. Methods: We performed whole genome sequencing (WGS) and whole exome sequencing (WES) on DNA isolated from peripheral blood from 36 KMT2A-r cases at time of remission. Sequencing was performed using an Illumina Hiseq 4000 or 2500 to a minimum depth of 90Gb (WGS) and 15Gb (WES). Alignment and variant calling were performed using the Dragon Bio-IT platform (v 3.2.8, Illumina). Blueprint Genetics clinical panels and the medical literature (Xa M et al., Nature 2018) were used to comprise a list of 346 genes associated with cancer predisposition and bone marrow failure syndromes. From this gene pool, variants were selected for analysis based on a variant allele frequency of ~50% and minor allele frequency Results: Of 351 variants initially identified, we found 3 likely pathogenic (LP) and 6 pathogenic(P) non-synonymous germline variants (for a total of 9 LP/P variants) and 144 variants of unknown significance (VUS). In total, 19.4% (n=7) of patient samples displayed at least one LP/P variant. Two patient samples contained 2 variants each. Variants classified as VUS, LP, or P were further characterized by possible causative pathway: 37.9% (n=58) of variants were in genes associated with bone marrow failure (BMF), 17.6% (n=27) in driver genes, 13.1% (n=20) in genes associated with inherited leukemias, 11.1% (n=17) in tumor suppressor genes, 7.8% (n=12) in tyrosine kinase genes, and 29.4% (n=45) in other predisposition genes . Many variants were present in more than one pathway and are represented as such. Table 1 demonstrates the genetic characteristics of the 9 P/LP variants found in our cohort. ERCC2 was the only gene with multiple LP/P variants across samples, accounting for 2 (1 LP, 1 P) of the 9 deleterious variants identified (22%). Conclusion: We identified germline variants in cancer predisposition genes in 19.4% of this cohort of infant ALL patients, a higher mutation rate than has previously been reported. Among pathways evaluated, variants in genes associated with bone marrow failure predisposition were the most frequent. Interestingly, variants in ERCC2, which encodes a protein involved with repair of damaged DNA, were recurrent among infants with KMT2A-r ALL. Future directions include comparison to germline variants in cancer predisposition genes in other infant and non-infant ALL cohorts. Disclosures Brown: Novartis: Membership on an entity's Board of Directors or advisory committees; Jazz: Membership on an entity's Board of Directors or advisory committees; Servier: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees. Guest:Syndax Pharmaceuticals: Consultancy.
- Published
- 2020
- Full Text
- View/download PDF
40. Recapitulation of human germline coding variation in an ultra-mutated infant leukemia
- Author
-
Parwinder Kaur, Alexander M. Gout, Ursula R. Kees, Catherine H. Cole, Rishi S. Kotecha, Jason Waithman, Charles S. Bond, Kim W. Carter, Mark N. Cruickshank, Bree Foley, and Abad A
- Subjects
Genetics ,0303 health sciences ,Mutation ,POLD1 ,Point mutation ,Somatic hypermutation ,Biology ,medicine.disease_cause ,Germline ,3. Good health ,Infant Acute Lymphoblastic Leukemia ,03 medical and health sciences ,0302 clinical medicine ,MSH2 ,030220 oncology & carcinogenesis ,medicine ,DNA mismatch repair ,030304 developmental biology - Abstract
BackgroundMixed lineage leukemia/Histone-lysine N-methyltransferase 2Agene rearrangements occur in 80% of infant acute lymphoblastic leukemia, but the role of cooperating events is unknown. While infant leukemias typically carry few somatic lesions, we identified a case with over 100 somatic point mutations per megabase and here report unique genomic-features of this case.ResultsThe patient presented at 82 days of age, one of the earliest manifestations of cancer hypermutation recorded. The transcriptional profile showed global similarities to canonical cases. Coding lesions were predominantly clonal and almost entirely targeting alleles reported in human genetic variation databases with a notable exception in the mismatch repair gene,MSH2. There were no rare germline alleles or somatic mutations affecting proof-reading polymerase genesPOLEorPOLD1, however there was a predicted damaging mutation in the error prone replicative polymerase,POLK. The patient’s diagnostic leukemia transcriptome was depleted of rare and low-frequency germline alleles due to loss-of-heterozygosity, while somatic point mutations targeted low-frequency and common human alleles in proportions that offset this discrepancy. Somatic signatures of ultra-mutations were highly correlated with germline single nucleotide polymorphic sites indicating a common role for 5-methylcytosine deamination, DNA mismatch repair and DNA adducts.ConclusionsThese data suggest similar molecular processes shaping population-scale human genome variation also underlies the rapid evolution of an infant ultra-mutated leukemia.
- Published
- 2018
- Full Text
- View/download PDF
41. Recent progress in the treatment of infant acute lymphoblastic leukemia
- Author
-
Daisuke Tomizawa
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,DOT1L ,Disease ,medicine.disease ,Lymphoma ,Infant Acute Lymphoblastic Leukemia ,Targeted therapy ,Leukemia ,hemic and lymphatic diseases ,Internal medicine ,Pediatrics, Perinatology and Child Health ,Immunology ,medicine ,Epigenetics ,business ,neoplasms - Abstract
Treatment of infants with acute lymphoblastic leukemia (ALL), especially those with mixed lineage leukemia (MLL) rearrangement (MLL-r), which account for approximately 80% of cases, is still a major challenge for pediatric hematologists and oncologists worldwide. Continuing efforts by collaborative clinical study groups in Europe, North America, and Japan have rescued approximately half of the MLL-r ALL patients with intensive chemotherapy with or without allogeneic hematopoietic stem cell transplantation. Recent progress has clarified the unique mechanism of MLL-r ALL: the aberrant methylation and histone modifications via DOT1L and other related molecules by MLL fusion proteins lead to leukemogenetic gene expression, thus to overt leukemia. In order to overcome this dismal subtype of ALL, novel targeted therapy based on leukemia biology is urgently needed. Due to the extreme rarity of the disease, collaboration between the study groups in Europe (Interfant), North America (Children's Oncology Group), and Japan (Japanese Pediatric Leukemia/Lymphoma Study Group) is under way.
- Published
- 2015
- Full Text
- View/download PDF
42. Outcome of relapsed infant acute lymphoblastic leukemia treated on the interfant-99 protocol
- Author
-
Francesco Locatelli, Ian Hann, Liisa Hovi, Martin Schrappe, Tomasz Szczepański, Emma M. C. Driessen, Lewis B. Silverman, Myriam Campbell, M. G. Valsecchi, Maria S. Felice, P De Lorenzo, Andrea Biondi, G Escherich, Rob Pieters, Thierry Leblanc, Alina Ferster, Georg Mann, J. Stary, Ram Suppiah, Jeffrey E. Rubnitz, Chi Kong Li, Ajay Vora, Driessen, E, de Lorenzo, P, Campbell, M, Felice, M, Ferster, A, Hann, I, Vora, A, Hovi, L, Escherich, G, Li, C, Mann, G, Leblanc, T, Locatelli, F, Biondi, A, Rubnitz, J, Schrappe, M, Silverman, L, Stary, J, Suppiah, R, Szczepanski, T, Valsecchi, M, Pieters, R, and Pediatrics
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pediatrics ,medicine.medical_treatment ,Treatment outcome ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,MIXED LINEAGE LEUKEMIA ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Chemotherapy ,Hematology ,business.industry ,Infant, Newborn ,Infant ,hemic and immune systems ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,3. Good health ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,MLL gene rearrangements ,Treatment Outcome ,Anesthesiology and Pain Medicine ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,030220 oncology & carcinogenesis ,Line (text file) ,ALL ,business ,030215 immunology - Abstract
Correction to: Leukemia (2016); 30(5), 1184–1187; doi:10.1038/leu.2015.246 Following the publication of this article the authors noted that the labels in Figures 1b and d have been switched. The correct labels in Figure 1b are; mixed lineage leukemia rearranged (MLL rearranged; dotted line) and unknown (thin solid line).
- Published
- 2015
- Full Text
- View/download PDF
43. Decreased induction morbidity and mortality following modification to induction therapy in infants with acute lymphoblastic leukemia enrolled on AALL0631: A report from the children's oncology group
- Author
-
Mignon L. Loh, Naomi J. Winick, Rob Pieters, Lillian Sung, Tamekia L. Jones, ZoAnn E. Dreyer, Cindy Y. Wang, Lia Gore, Meenakshi Devidas, William L. Carroll, Paola De Lorenzo, Joanne M. Hilden, Maria Grazia Valsecchi, Patrick A. Brown, Elizabeth A. Raetz, Wanda L. Salzer, and Stephen P. Hunger
- Subjects
Pediatrics ,medicine.medical_specialty ,Poor prognosis ,Hematology ,business.industry ,Lymphoblastic Leukemia ,Case-control study ,Induction chemotherapy ,Infant Acute Lymphoblastic Leukemia ,Oncology ,hemic and lymphatic diseases ,Induction therapy ,Internal medicine ,Pediatrics, Perinatology and Child Health ,medicine ,business ,Cohort study - Abstract
Background Infants with acute lymphoblastic leukemia (ALL) have a poor prognosis. Intensification of therapy has resulted in fewer relapses but increased early deaths, resulting in failure to improve survival.
- Published
- 2014
- Full Text
- View/download PDF
44. Clonal variegation and dynamic competition of leukemia-initiating cells in infant acute lymphoblastic leukemia with MLL rearrangement
- Author
-
Lilian Wittmann, L Corral, Michela Bardini, L Lo Nigro, Giovanni Cazzaniga, Zhi Ma, Giuseppe Basso, Sten Eirik W. Jacobsen, Petter S. Woll, Andrea Biondi, Sidinh Luc, Bardini, M, Woll, P, Corral, L, Luc, S, Wittmann, L, Ma, Z, Lo Nigro, L, Basso, G, Biondi, A, Cazzaniga, G, and Jacobsen, S
- Subjects
Cancer Research ,Clone (cell biology) ,Biology ,Immunophenotyping ,Mice ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,In Situ Hybridization, Fluorescence ,Variegation ,Gene Rearrangement ,T-cell receptor ,Infant ,Histone-Lysine N-Methyltransferase ,Hematology ,Gene rearrangement ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Phenotype ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,Oncology ,Leukemia-initiating cells, Infant leukemia, MLL gene rearrangement ,Immunology ,Heterografts ,Myeloid-Lymphoid Leukemia Protein - Abstract
Distinct from other forms of acute lymphoblastic leukemia (ALL), infant ALL with mixed lineage leukemia (MLL) gene rearrangement, the most common leukemia occurring within the first year of life, might arise without the need for cooperating genetic lesions. Through Ig/TCR rearrangement analysis of MLL-AF4+ infant ALL at diagnosis and xenograft leukemias from mice transplanted with the same diagnostic samples, we established that MLL-AF4+ infant ALL is composed of a branching subclonal architecture already at diagnosis, frequently driven by an Ig/TCR-rearranged founder clone. Some MLL-AF4+ clones appear to be largely quiescent at diagnosis but can reactivate and dominate when serially transplanted into immunodeficient mice, whereas other dominant clones at diagnosis can become more quiescent, suggesting a dynamic competition between actively proliferating and quiescent subclones. Investigation of paired diagnostic and relapse samples suggested that relapses often occur from subclones already present but more quiescent at diagnosis. Copy-number alterations identified at relapse might contribute to the activation and expansion of previously quiescent subclones. Finally, each of the identified subclones is able to contribute to the diverse phenotypic pool of MLL-AF4+ leukemia-propagating cells. Unraveling of the subclonal architecture and dynamics in MLL+ infant ALL may provide possible explanations for the therapy resistance and frequent relapses observed in this group of poor prognosis ALL.
- Published
- 2014
- Full Text
- View/download PDF
45. Infants with Cancer: A Unique Population
- Author
-
Wen-Ming Hsu, Yen Lin Liu, Shu Huey Chen, and James S. Miser
- Subjects
Pediatrics ,medicine.medical_specialty ,education.field_of_study ,business.industry ,medicine.medical_treatment ,Population ,Cancer ,General Medicine ,Disease ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,Radiation therapy ,Etiology ,Medicine ,Germ cell tumors ,business ,education ,Cancer Etiology - Abstract
Cancer occurring in infants often has a clinical and biological behavior that is different from cancers occurring in older children. The histological distribution of cancers in infants is different from that in older children. The five most common types of cancer occurring in infants in Taiwan are leukemia, neuroblastoma, germ cell tumors, central nervous system neoplasms, and retinoblastomas. Cancer in infants represents a unique situation in which to study cancer etiology. A significant number of infants with cancer have a genetic susceptibility to the disease; however, some emerging studies suggest a potential role for environmental, dietary, and drug exposures in the etiology of infant cancers. Further definitive trials will be necessary to establish clear associations, however. Because of their very young age and the immaturity of many of their physiological systems, the approach to treatment in young infants differs from that in older children. The infant's response to treatment also differs from older children, indicating the unique biological properties of cancer in infants that may explain different clinical outcomes in this unique population. Infant acute lymphoblastic leukemia has a much inferior outcome compared with the outcome in older children. In contrast, neuroblastoma in infants has a superior outcome to that seen in older children. The care of infants with cancer is extremely challenging as a result of their increased vulnerability to the acute complications associated with intensive, multimodal treatment and to the long-term sequelae of chemotherapy and radiotherapy on the rapidly growing and developing infant. The chemotherapy-related toxicity seen in very young infants of less than 3 months of age may be due to a lower total body water content, lower P450 enzyme activity, lower serum protein binding, and immature renal function. The high susceptibility of immature tissues to radiotherapy-induced damage has led to the delivery of radiotherapy being limited in this age group. Protocols specific to the characteristics of this population are currently yielding promising results.
- Published
- 2014
- Full Text
- View/download PDF
46. KMT2A-Rearranged Infant Acute Lymphoblastic Leukemia Cells Undergo ER-Stress-Induced Apoptosis Following Exposure to Proteasome Inhibitors
- Author
-
Tanja A. Gruber, Cary Koss, and Wonil Kim
- Subjects
business.industry ,Bortezomib ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Infant Acute Lymphoblastic Leukemia ,Leukemia ,Proteasome ,Acute lymphocytic leukemia ,medicine ,Unfolded protein response ,Proteasome inhibitor ,Cancer research ,business ,Multiple myeloma ,medicine.drug - Abstract
Infants diagnosed with KMT2A-rearranged (KMT2Ar) acute lymphoblastic leukemia (ALL) have a poor prognosis with an event free survival of 23-44%. To identify new treatment approaches we previously performed in vitro and in vivo assays to evaluate the activity of FDA approved compounds in 15 primary KMT2Ar infant leukemia samples. Three classes of agents were found to be active in these assays: proteasome inhibitors, anthracyclines, and histone deacetylase inhibitors (HDACi). KMT2Ar infant leukemia samples were exquisitely sensitive to the proteasome inhibitor bortezomib, requiring 10-100 fold less drug to achieve 50% toxicity when compared to non-KMT2Ar childhood ALL. Bortezomib is FDA approved for multiple myeloma and laboratory studies using this model system have previously demonstrated responses to be mediated through several mechanisms including NFKB inhibition, stabilization of cell cycle regulatory proteins, and perhaps most importantly the induction of an unfolded protein response (UPR) and endoplasmic reticulum (ER)-stress-induced apoptosis. To evaluate global protein dynamics in KMT2Ar ALL cells treated with bortezomib, we performed tandem mass tag (TMT) quantitative mass spectrometry on synchronized SEM cells exposed to either 50nM of bortezomib or DMSO at 0 hours (hr), 6hr, 12hr, 16hr, and 20hr. Applying pairwise comparison for 9232 unique proteins measured over the time course compared to untreated controls, we identified 1593 proteins with a log2 fold change >1.5 in bortezomib treated cells compared to 101 proteins in the DMSO control (FDR While GADD34 has been shown to be the main phosphatase that functions in a negative feedback loop to resolve cell stress, recent data suggests that stabilization of CReP mRNA by ER stress is able to reverse eIF2α phosphorylation at later stages of UPR leading to re-expression of key UPR proteins. Further, p-eIF2α-attenuated protein synthesis, and not ATF4 mRNA translation has been shown to promote cell survival. Our data support a model whereby the UPR and ER-stress in KMT2Ar ALL cells is induced upon exposure to bortezomib leading to increased levels of ATF4 and CHOP. Attenuation of p-eIF2α by CReP further contributes to cell death through the recovery of protein synthesis in a setting of limited protein folding capacity. These results support the use of proteasome inhibitors in KMT2Ar leukemia which is currently being formally evaluated in a Phase II clinical trial for newly diagnosed patients with infant ALL (NCT02553460). Disclosures Gruber: Bristol-Myers Squibb: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
47. Late Effects in Survivors of Infant Acute Lymphoblastic Leukemia from the 3 Consecutive Japanese Nationwide Clinical Trials
- Author
-
Miho Maeda, Hisamichi Tauchi, Keizo Horibe, Katsuyoshi Koh, Akiko Saito, Akira Hayakawa, Atsushi Manabe, Yuki Aoki, Akiko Kada, Eiichi Ishii, Kazutoshi Koike, Daisuke Tomizawa, Yasushi Ishida, Shuki Mizutani, and Takako Miyamura
- Subjects
Pediatrics ,medicine.medical_specialty ,Childhood leukemia ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Infant Acute Lymphoblastic Leukemia ,Clinical trial ,Transplantation ,Regimen ,Leukemia ,Acute lymphocytic leukemia ,medicine ,business ,Busulfan ,medicine.drug - Abstract
Background Very few studies have reported the incidence of and risk factor for late effects of infant leukemia. In addition, the long-term impact of a preparative regimen for very young children undergoing allogeneic stem cell transplantation (SCT) is not evident. Method To examine the late effects in survivors of infant acute lymphoblastic leukemia (ALL) treated on the Japanese Infant Leukemia Study Group (JILSG) trials MLL96, MLL98 and the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) trial MLL03, we performed a cross-sectional survey using questionnaires by the attending pediatricians from 2015 to 2017. In all 3 studies, infants with KMT2A gene rearranged (MLL-R) ALL were allocated to receive SCT in their first remission. Choice of preparative regimen was left to the physician's choice in the MLL96/98 studies, while use of non-total body irradiation (TBI) regimen with busulfan (BU) was mandatory in the MLL03 study. Results Seventy out of 102 (69%) eligible subjects were evaluated: 33 were males and 37 were females. 62 patients had an MLL-R ALL. The median age of diagnosis was 5.5 months (range, 0-12m) and 35 (50%) were Conclusion In this study, survivors of infant leukemia had prolonged and various types of late effects. We observed serious growth failure in height and weight in patients treated with SCT and it was further pronounced in the TBI group and in the multiple SCT (BU+TBI) group. More than half of the patients had dental late effects and a third of all the patients had thyroid dysfunctions, which were more frequent in children diagnosed earlier. The number of patients over 20 years was limited in our study, more complications may increase in additional follow-up. Continuous long-term follow up and optimal intervention are crucial for survivors of infant leukemia who are transplanted. Disclosures Kada: Bayer Yakuhin: Other: personal fees for a member of independent data monitoring committee of clinical trials, outside of the submitted work..
- Published
- 2019
- Full Text
- View/download PDF
48. FLT3 Inhibitor Correlative Laboratory Assays Impact Outcomes in KMT2A-Rearranged Infant Acute Lymphoblastic Leukemia (ALL) Patients Treated with Lestaurtinib: AALL0631, a Children's Oncology Group Study
- Author
-
Mignon L. Loh, John A. Kairalla, William L. Carroll, Patrick A. Brown, Donald Small, Stephen P. Hunger, Michael J. Borowitz, Meenakshi Devidas, Cindy Wang, ZoAnn E. Dreyer, Joanne M. Hilden, Andrew J. Carroll, Elizabeth A. Raetz, and Nyla A. Heerema
- Subjects
Oncology ,medicine.medical_specialty ,Lestaurtinib ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Infant Acute Lymphoblastic Leukemia ,Transplantation ,Leukemia ,Tolerability ,Internal medicine ,Acute lymphocytic leukemia ,Pharmacodynamics ,medicine ,business ,medicine.drug - Abstract
Background/Objectives Infants with ALL have poor outcomes, especially the 70-80% with KMT2A rearrangements (KMT2A-r). Intensification of chemotherapy has reached the limit of tolerability, and stem cell transplantation (SCT) has not improved outcomes. The FLT3 kinase is overexpressed and constitutively activated in KMT2A-r ALL. FLT3 inhibitors (FLT3i) selectively kill KMT2A-r ALL cells in vitro and in vivo, and synergize with chemotherapy. COG AALL0631 was a randomized trial that tested the hypothesis that the addition of the FLT3i lestaurtinib to post-induction chemotherapy would improve outcomes for KMT2A-r ALL.As previously reported, the addition of lestaurtinib did not improve outcomes [3-year disease-free/overall survival (DFS/OS) 39%/46% without lestaurtinib vs. 37%/45% with lestaurtinib, log-rank p=0.78/0.92]. We hypothesized that correlative laboratory measures of FLT3i pharmacodynamics and ex vivo FLT3i sensitivity would enhance our understanding of the clinical responses for KMT2A-r infants treated with lestaurtinib. Design/Methods FLT3i pharmacodynamics (PD) were determined using plasma inhibitory activity (PIA) measured at 5 trough time points during lestaurtinib treatment, using Western blotting and densitometry (normalized to pre-treatment levels). PIA ranged from 0% (no evidence of FLT3 inhibition) to 100% (full inhibition). Mean PIA of 5 troughs was calculated for each patient, and was categorized based on pre-defined protocol criteria as "adequate", or FLT3 inhibited (≥85%) vs "inadequate", or FLT3 uninhibited ( Results Of 210 eligible patients, 146 were KMT2A-r, and 78 were assigned to receive lestaurtinib. Of these, data were available for FLT3i PD (n=73), FLT3i EVS (n=62), MRD (n=55), and DFS/OS (n=78). FLT3i PD and FLT3i EVS significantly correlated with both survival and MRD. For FLT3i PD, 27 were "FLT3 inhibited" and 46 were "FLT3 uninhibited". The DFS/OS for inhibited was 59%/67% vs. 28%/37% for uninhibited (p=0.01/0.02). The week 10 MRD-negative rate was 90% for inhibited vs. 52% for uninhibited (p=0.003). For FLT3i EVS, of the 62 lestaurtinib-treated patients with data, 42 were "sensitive" and 20 were "resistant". The DFS/OS for sensitive was 52%/62% vs. 5%/11% for resistant (p Conclusion While the addition of lestaurtinib to chemotherapy did not improve outcomes, patients achieving potent pharmacodynamic inhibition of FLT3 and those whose leukemia cells were sensitive to ex-vivo FLT3-inhibitor induced cytotoxicity did benefit from the addition of lestaurtinib. Selection of patients with FLT3-sensitive leukemia, real-time intra-patient dose escalation of patients with inadequate pharmacodynamics, and/or using a more potent FLT3 inhibitor may enhance the efficacy of FLT3 inhibition for KMT2A-r infant ALL. Disclosures Raetz: Pfizer: Research Funding. Loh:Medisix Therapeutics, Inc.: Membership on an entity's Board of Directors or advisory committees. Hunger:Bristol Myers Squibb: Consultancy; Amgen: Consultancy, Equity Ownership; Novartis: Consultancy; Jazz: Honoraria. Borowitz:Beckman Coulter: Honoraria. Small:Pharos I, B & T: Consultancy, Research Funding; InSilico Medicine: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2019
- Full Text
- View/download PDF
49. Identification of hub genes and molecular mechanisms in infant acute lymphoblastic leukemia withMLLgene rearrangement
- Author
-
Zijian Li, Juan Cheng, Yaming Xi, and Hao Zhang
- Subjects
lcsh:Medicine ,Acute lymphoblastic leukemia ,PTPRC ,General Biochemistry, Genetics and Molecular Biology ,Protein kinase C signaling ,03 medical and health sciences ,0302 clinical medicine ,Gene interaction ,Neuron projection extension ,hemic and lymphatic diseases ,Mixed-lineage leukemia ,KEGG ,030304 developmental biology ,Genetics ,0303 health sciences ,biology ,General Neuroscience ,lcsh:R ,Infant ,General Medicine ,Gene rearrangement ,Infant Acute Lymphoblastic Leukemia ,Gene expression profiles ,030220 oncology & carcinogenesis ,Differentially expressed genes ,biology.protein ,General Agricultural and Biological Sciences ,Extracellular matrix organization - Abstract
Infant acute lymphoblastic leukemia (ALL) with the mixed lineage leukemia (MLL) gene rearrangement (MLL-R) is considered a distinct leukemia from childhood or non-MLL-R infant ALL. To detect key genes and elucidate the molecular mechanisms ofMLL-R infant ALL, microarray expression data were downloaded from the Gene Expression Omnibus (GEO) database, and differentially expressed genes (DEGs) betweenMLL-R and non-MLL-R infant ALL were identified. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were carried out. Then, we constructed a protein-protein interaction (PPI) network and identified the hub genes. Finally, drug-gene interactions were mined. A total of 139 cases ofMLL-R infant ALL including 77 (55.4%) fusions withAF4, 38 (27.3%) withENL, 14 (10.1%) withAF9, and 10 (7.2%) other gene fusions were characterized. A total of 236 up-regulated and 84 down-regulated DEGs were identified. The up-regulated DEGs were mainly involved in homophilic cell adhesion, negative regulation of apoptotic process and cellular response to drug GO terms, while down-regulated DEGs were mainly enriched in extracellular matrix organization, protein kinase C signaling and neuron projection extension GO terms. The up-regulated DEGs were enriched in seven KEGG pathways, mainly involving transcriptional regulation and signaling pathways, and down-regulated DEGs were involved in three main KEGG pathways including Alzheimer’s disease, TGF-beta signaling pathway, and hematopoietic cell lineage. The PPI network included 297 nodes and 410 edges, withMYC,ALB,CD44,PTPRCandTNFidentified as hub genes. Twenty-three drug-gene interactions including four up-regulated hub genes and 24 drugs were constructed by Drug Gene Interaction database (DGIdb). In conclusion,MYC,ALB,CD44,PTPRCandTNFmay be potential bio-markers for the diagnosis and therapy ofMLL-R infant ALL.
- Published
- 2019
- Full Text
- View/download PDF
50. Abstract 3649: Allelic imbalance in KMT2A-rearranged infant acute lymphoblastic leukemia
- Author
-
Neil A. Miller, Margaret Gibson, Rumen Kostadinov, Shannon Kelley, Bing Ge, Midhat S. Farooqi, Tomi Pastinen, Patrick A. Brown, Erin M. Guest, Warren A. Cheung, Byunggil Yoo, and Emily G. Farrow
- Subjects
Genetics ,Whole genome sequencing ,Cancer Research ,biology ,Cancer ,Single-nucleotide polymorphism ,medicine.disease ,Infant Acute Lymphoblastic Leukemia ,KMT2A ,Oncology ,Allelic Imbalance ,biology.protein ,medicine ,Allele ,1000 Genomes Project - Abstract
Background Infant acute lymphoblastic leukemia (ALL) is a malignant disorder with poor clinical outcome. It is well-known that infant ALL cases with rearrangement of the KMT2A gene (KMT2A-r) have an even poorer prognosis than non-KMT2A-r cases. Interestingly, KMT2A-r infant ALL cases have remarkably few other genetic alterations. We hypothesized that non-coding events in cancer genomes (e.g. loss of expression or methylation) may play a role in this disease. Such events can be captured using genomic analyses in haploid genomes using analytical approaches for allelic imbalance quantification. Here, we examine whether allelic imbalance is a feature of infant ALL. Methods We performed whole genome sequencing (WGS) and RNA sequencing on peripheral blood or bone marrow specimens from 29 KMT2A-r cases and 14 non-KMT2A-r cases at diagnosis (DX), remission (MD), and relapse (RL) as applicable. WGS data from MD samples was phased using a 1000 Genomes reference panel. Biallelic expression was measured on the phased genome for transcripts with at least 2 SNPs and at least 15 aligned reads. Lesser allele fraction (LAF; allele fraction for the less prevalent allele) for DX/RL versus MD samples was compared for transcripts that had LAFs available in at least 3 cases using t-test. Transcript-level p-values were aggregated at the gene level using the Sidak method. Results Allelic imbalance in expression (LAF Discussion Our study suggests that allelic imbalance quantification may help uncover novel molecular mechanisms in infant ALL, especially KMT2A-r cases. However, similar to gene expression, the patterns of allelic imbalance in KMT2A-r cases at DX do not allow efficient prediction of which patients go on to relapse. Citation Format: Byunggil Yoo, Midhat S. Farooqi, Rumen Kostadinov, Warren Cheung, Emily Farrow, Shannon Kelley, Neil Miller, Bing Ge, Margaret Gibson, Patrick Brown, Erin M. Guest, Tomi Pastinen. Allelic imbalance in KMT2A-rearranged infant acute lymphoblastic leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3649.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.