5,365 results on '"Childhood Leukemia"'
Search Results
2. A dual-role for IL-10: From leukemogenesis to the tumor progression in acute lymphoblastic leukemia
- Author
-
Silva, Flavio Souza, Barros-Lima, Amanda, Souza-Barros, Mateus, Crespo-Neto, Juniel Assis, Santos, Vitória Giovanna Rodrigues, Pereira, Daniele Sá, Alves-Hanna, Fabíola Silva, Magalhães-Gama, Fábio, Faria, Jerusa Araújo Quintão Arantes, and Costa, Allyson Guimarães
- Published
- 2023
- Full Text
- View/download PDF
3. Titrating chimeric antigen receptors on CAR T cells enabled by a microfluidic-based dosage-controlled intracellular mRNA delivery platform
- Author
-
Chen, Yu-Hsi, Mirza, Mahnoor, Jiang, Ruoyu, and Lee, Abraham P
- Subjects
Fluid Mechanics and Thermal Engineering ,Engineering ,Gene Therapy ,Pediatric Cancer ,Biotechnology ,Immunotherapy ,Pediatric ,Rare Diseases ,Orphan Drug ,Genetics ,Bioengineering ,Childhood Leukemia ,Hematology ,Cancer ,5.2 Cellular and gene therapies ,5.1 Pharmaceuticals ,Classical Physics ,Interdisciplinary Engineering ,Nanotechnology ,Nanoscience & Nanotechnology ,Fluid mechanics and thermal engineering - Abstract
Chimeric antigen receptor (CAR) T-cell therapy shows unprecedented efficacy for cancer treatment, particularly in treating patients with various blood cancers, most notably B-cell acute lymphoblastic leukemia. In recent years, CAR T-cell therapies have been investigated for treating other hematologic malignancies and solid tumors. Despite the remarkable success of CAR T-cell therapy, cytokine release syndrome (CRS) is an unexpected side effect that is potentially life-threatening. Our aim is to reduce pro-inflammatory cytokine release associated with CRS by controlling CAR surface density on CAR T cells. We show that CAR expression density can be titrated on the surface of primary T cells using an acoustic-electric microfluidic platform. The platform performs dosage-controlled delivery by uniformly mixing and shearing cells, delivering approximately the same amount of CAR gene coding mRNA into each T cell.
- Published
- 2024
4. Pooled analysis of recent studies of magnetic fields and childhood leukemia
- Author
-
Amoon, Aryana T., Swanson, John, Magnani, Corrado, Johansen, Christoffer, and Kheifets, Leeka
- Published
- 2022
- Full Text
- View/download PDF
5. Early-life tobacco exposure is causally implicated in aberrant RAG-mediated recombination in childhood acute lymphoblastic leukemia
- Author
-
Liu, Tanxin, Xu, Keren, Pardeshi, Anmol, Myint, Swe Swe, Kang, Alice Y, Morimoto, Libby M, Lieber, Michael R, Wiemels, Joseph L, Kogan, Scott C, Metayer, Catherine, and de Smith, Adam J
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Clinical Sciences ,Oncology and Carcinogenesis ,Tobacco Smoke and Health ,Childhood Leukemia ,Hematology ,Cancer ,Rare Diseases ,Tobacco ,Pediatric ,Pediatric Cancer ,Good Health and Well Being ,Immunology ,Cardiovascular medicine and haematology ,Clinical sciences ,Oncology and carcinogenesis - Published
- 2024
6. Folate metabolism and risk of childhood acute lymphoblastic leukemia: a genetic pathway analysis from the Childhood Cancer and Leukemia International Consortium
- Author
-
Metayer, Catherine, Spector, Logan G, Scheurer, Michael E, Jeon, Soyoung, Scott, Rodney J, Takagi, Masatoshi, Clavel, Jacqueline, Manabe, Atsushi, Ma, Xiaomei, Hailu, Elleni M, Lupo, Philip J, Urayama, Kevin Y, Bonaventure, Audrey, Kato, Motohiro, Meirhaeghe, Aline, Chiang, Charleston WK, Morimoto, Libby M, and Wiemels, Joseph L
- Subjects
Epidemiology ,Health Sciences ,Pediatric Cancer ,Cancer ,Clinical Research ,Rare Diseases ,Childhood Leukemia ,Human Genome ,Hematology ,Pediatric ,Genetics ,Minority Health ,Health Disparities ,2.1 Biological and endogenous factors ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Folic Acid ,Polymorphism ,Single Nucleotide ,Child ,Case-Control Studies ,Female ,Male ,Genome-Wide Association Study ,Risk Factors ,Genetic Predisposition to Disease ,Child ,Preschool ,Medical and Health Sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundPrenatal folate supplementation has been consistently associated with a reduced risk of childhood acute lymphoblastic leukemia (ALL). Previous germline genetic studies examining the one carbon (folate) metabolism pathway were limited in sample size, scope, and population diversity and led to inconclusive results.MethodsWe evaluated whether ∼2,900 single-nucleotide polymorphisms (SNP) within 46 candidate genes involved in the folate metabolism pathway influence the risk of childhood ALL, using genome-wide data from nine case-control studies in the Childhood Cancer and Leukemia International Consortium (n = 9,058 cases including 4,510 children of European ancestry, 3,018 Latinx, and 1,406 Asians, and 92,364 controls). Each study followed a standardized protocol for quality control and imputation of genome-wide data and summary statistics were meta-analyzed for all children combined and by major ancestry group using METAL software.ResultsNone of the selected SNPs reached statistical significance, overall and for major ancestry groups (using adjusted Bonferroni P-value of 5 × 10-6 and less-stringent P-value of 3.5 × 10-5 accounting for the number of "independent" SNPs). None of the 10 top (nonsignificant) SNPs and corresponding genes overlapped across ancestry groups.ConclusionsThis large meta-analysis of original data does not reveal associations between many common genetic variants in the folate metabolism pathway and childhood ALL in various ancestry groups.ImpactGenetic variants in the folate pathway alone do not appear to substantially influence childhood acute lymphoblastic leukemia risk. Other mechanisms such as gene-folate interaction, DNA methylation, or maternal genetic effects may explain the observed associations with self-reported prenatal folate intake.
- Published
- 2024
7. Residential proximity to plant nurseries and risk of childhood leukemia
- Author
-
Nguyen, Andrew, Crespi, Catherine M., Vergara, Ximena, Chun, Nicholas, and Kheifets, Leeka
- Published
- 2021
- Full Text
- View/download PDF
8. LNK/SH2B3 as a novel driver in juvenile myelomonocytic leukemia
- Author
-
Wintering, Astrid, Hecht, Anna, Meyer, Julia, Wong, Eric B, Hübner, Juwita, Abelson, Sydney, Feldman, Kira, Kennedy, Vanessa E, Peretz, Cheryl AC, French, Deborah L, Maguire, Jean Ann, Jobaliya, Chintan, Vasquez, Marta Rojas, Desai, Sunil, Dulman, Robin, Nemecek, Eneida, Haines, Hilary, Hammad, Mahmoud, El Haddad, Alaa, Kogan, Scott C, Abdullaev, Zied, Chehab, Farid F, Tasian, Sarah K, Smith, Catherine C, Loh, Mignon L, and Stieglitz, Elliot
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Cancer ,Childhood Leukemia ,Hematology ,Pediatric Cancer ,Stem Cell Research - Nonembryonic - Human ,Stem Cell Research - Nonembryonic - Non-Human ,Genetics ,Pediatric ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Rare Diseases ,2.1 Biological and endogenous factors ,Humans ,Leukemia ,Myelomonocytic ,Juvenile ,Adaptor Proteins ,Signal Transducing ,Male ,Female ,Infant ,Child ,Preschool ,Mutation ,Intracellular Signaling Peptides and Proteins ,Child ,Signal Transduction ,Pyrazoles ,Nitriles ,Pyrimidines ,Immunology - Abstract
Mutations in five canonical Ras pathway genes (NF1, NRAS, KRAS, PTPN11 and CBL) are detected in nearly 90% of patients with juvenile myelomonocytic leukemia (JMML), a frequently fatal malignant neoplasm of early childhood. In this report, we describe seven patients diagnosed with SH2B3-mutated JMML, including five patients who were found to have initiating, loss-of-function mutations in the gene. SH2B3 encodes the adaptor protein LNK, a negative regulator of normal hematopoiesis upstream of the Ras pathway. These mutations were identified to be germline, somatic or a combination of both. Loss of function of LNK, which has been observed in other myeloid malignancies, results in abnormal proliferation of hematopoietic cells due to cytokine hypersensitivity and activation of the JAK/STAT signaling pathway. In vitro studies of induced pluripotent stem cell-derived JMML-like hematopoietic progenitor cells also demonstrated sensitivity of SH2B3-mutated hematopoietic progenitor cells to JAK inhibition. Lastly, we describe two patients with JMML and SH2B3 mutations who were treated with the JAK1/2 inhibitor ruxolitinib. This report expands the spectrum of initiating mutations in JMML and raises the possibility of targeting the JAK/STAT pathway in patients with SH2B3 mutations.
- Published
- 2024
9. Early-life tobacco exposure is causally implicated in aberrant RAG-mediated recombination in childhood acute lymphoblastic leukemia
- Author
-
de Smith, Adam, Liu, Tanxin, Xu, Keren, Pardeshi, Anmol, Myint, Swe Swe, Kang, Alice, Morimoto, Libby, Lieber, Michael, Wiemels, Joseph, Kogan, Scott, and Metayer, Catherine
- Subjects
Biomedical and Clinical Sciences ,Public Health ,Health Sciences ,Pediatric Cancer ,Cancer Genomics ,Pediatric ,Hematology ,Cancer ,Rare Diseases ,Prevention ,Genetics ,Human Genome ,Childhood Leukemia ,Tobacco Smoke and Health ,Biotechnology ,Tobacco ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Good Health and Well Being - Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in children, yet few environmental risk factors have been identified. We previously found an association between early-life tobacco smoke exposure and frequency of somatic deletions of 8 leukemia driver genes among childhood ALL patients in the California Childhood Leukemia Study. To expand analysis genome-wide and examine potential mechanisms, we conducted tumor whole-genome sequencing in 35 ALL patients, including 18 with high prenatal tobacco exposure and 17 with low exposure as determined by established epigenetic biomarkers. High tobacco exposure patients had significantly more structural variants (P < .001) and deletions (P = .001) genome-wide than low exposure patients. Investigation of off-target RAG recombination revealed that 41% of deletions in the high tobacco exposure patients were putatively RAG-mediated (full RAG motif identified at one or both breakpoints) compared with only 21% in the low exposure group (P = .001). In a multilevel model, deletions in high tobacco exposure patients were 2.44-fold (95% CI:1.13-5.38) more likely to be putatively RAG-mediated than deletions in low exposure patients. No point mutational signatures were associated with prenatal tobacco exposure. Our findings suggest that early-life tobacco smoke exposure may promote leukemogenesis by driving development of somatic deletions in pre-leukemic lymphocytes via off-target RAG recombination.
- Published
- 2024
10. Efficacy of the Allosteric MEK Inhibitor Trametinib in Relapsed and Refractory Juvenile Myelomonocytic Leukemia: a Report from the Children's Oncology Group.
- Author
-
Stieglitz, Elliot, Lee, Alex G, Angus, Steven P, Davis, Christopher, Barkauskas, Donald A, Hall, David, Kogan, Scott C, Meyer, Julia, Rhodes, Steven D, Tasian, Sarah K, Xuei, Xiaoling, Shannon, Kevin, Loh, Mignon L, Fox, Elizabeth, and Weigel, Brenda J
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Oncology and Carcinogenesis ,Clinical Research ,Hematology ,Cancer ,Clinical Trials and Supportive Activities ,Regenerative Medicine ,Transplantation ,Pediatric ,Pediatric Cancer ,Childhood Leukemia ,Rare Diseases ,Stem Cell Research ,Evaluation of treatments and therapeutic interventions ,6.2 Cellular and gene therapies ,6.1 Pharmaceuticals ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Juvenile myelomonocytic leukemia (JMML) is a hematologic malignancy of young children caused by mutations that increase Ras signaling output. Hematopoietic stem cell transplantation (HSCT) is a potentially curative treatment, but patients with relapsed or refractory (advanced) disease have dismal outcomes. This phase II trial evaluated the safety and efficacy of trametinib, an oral MEK1/2 inhibitor, in patients with advanced JMML. Ten infants and children were enrolled, and the objective response rate was 50%. Four patients with refractory disease proceeded to HSCT after receiving trametinib. Three additional patients completed all 12 cycles permitted on study and continue to receive off-protocol trametinib without HSCT. The remaining three patients had progressive disease with two demonstrating molecular evolution by the end of cycle 2. Transcriptomic and proteomic analyses provided novel insights into the mechanisms of response and resistance to trametinib in JMML. ClinicalTrials.gov Identifier: NCT03190915. Significance: Trametinib was safe and effective in young children with relapsed or refractory JMML, a lethal disease with poor survival rates. Seven of 10 patients completed the maximum 12 cycles of therapy or used trametinib as a bridge to HSCT and are alive with a median follow-up of 24 months.
- Published
- 2024
11. Maternal autoimmune disease and its association with childhood cancer: A population-based case-control study in Denmark
- Author
-
Orimoloye, Helen T, Nguyen, Nicholas, Deng, Chuanjie, Saechao, Chai, Ritz, Beate, Olsen, Jorn, Hansen, Johnni, and Heck, Julia E
- Subjects
Epidemiology ,Biomedical and Clinical Sciences ,Health Sciences ,Oncology and Carcinogenesis ,Autoimmune Disease ,Cancer ,Brain Disorders ,Hematology ,Childhood Leukemia ,Brain Cancer ,Neurosciences ,Pediatric ,Rare Diseases ,Pediatric Cancer ,2.1 Biological and endogenous factors ,2.4 Surveillance and distribution ,Good Health and Well Being ,Maternal autoimmune diseases ,childhood cancer ,inflammatory bowel disease ,leukemia ,pregnancy ,psoriasis ,rheumatoid arthritis - Abstract
BackgroundAutoimmune diseases have been linked to an increased risk of pregnancy-related complications. A family history of autoimmune diseases may be related to the risk of childhood cancer based on similar histocompatibility antigens. We utilized data from national registries in Denmark to examine associations between maternal autoimmune disease and cancer in their offspring.MethodsWe linked data from several national registries in Denmark to identify childhood cancer cases in children
- Published
- 2024
12. A noncoding regulatory variant in IKZF1 increases acute lymphoblastic leukemia risk in Hispanic/Latino children
- Author
-
de Smith, Adam J, Wahlster, Lara, Jeon, Soyoung, Kachuri, Linda, Black, Susan, Langie, Jalen, Cato, Liam D, Nakatsuka, Nathan, Chan, Tsz-Fung, Xia, Guangze, Mazumder, Soumyaa, Yang, Wenjian, Gazal, Steven, Eng, Celeste, Hu, Donglei, Burchard, Esteban González, Ziv, Elad, Metayer, Catherine, Mancuso, Nicholas, Yang, Jun J, Ma, Xiaomei, Wiemels, Joseph L, Yu, Fulong, Chiang, Charleston WK, and Sankaran, Vijay G
- Subjects
Biological Sciences ,Genetics ,Minority Health ,Childhood Leukemia ,Cancer ,Hematology ,Pediatric Cancer ,Health Disparities ,Pediatric ,Rare Diseases ,Human Genome ,Humans ,Child ,Genetic Predisposition to Disease ,Polymorphism ,Single Nucleotide ,Transcription Factors ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Hispanic or Latino ,Ikaros Transcription Factor ,B-ALL ,GWAS ,IKZF1 ,Indigenous American ,acute lymphoblastic leukemia ,cancer disparity ,cancer predisposition ,childhood leukemia ,fine-mapping ,hematopoiesis - Abstract
Hispanic/Latino children have the highest risk of acute lymphoblastic leukemia (ALL) in the US compared to other racial/ethnic groups, yet the basis of this remains incompletely understood. Through genetic fine-mapping analyses, we identified a new independent childhood ALL risk signal near IKZF1 in self-reported Hispanic/Latino individuals, but not in non-Hispanic White individuals, with an effect size of ∼1.44 (95% confidence interval = 1.33-1.55) and a risk allele frequency of ∼18% in Hispanic/Latino populations and
- Published
- 2024
13. Radiation-induced meningiomas following childhood cranial radiotherapy—report of four cases.
- Author
-
Ay, Larisa Andrada, Ozgiray, Erkin, Akintürk, Nevhis, Kamer, Emine Serra, and Ertan, Yeşim
- Abstract
Background: Radiation-induced meningiomas represent a late side effect of cranial radiation therapy. The most widely used diagnostic criteria, despite the lack of a standard description, is a meningioma that develops in a previously irradiated area following a predetermined time interval. Case presentation: We present four cases of late-onset cranial atypical meningiomas following childhood cranial radiotherapy. Four patients (two females and two males) between 4 and 13 years of age at the time of cranial irradiation developed secondary intracranial meningioma after a latency period of 8–34 years. Surgical resection of the tumors was performed, and histopathological examination revealed a diagnosis of atypical meningioma in all four patients. Conclusion: Radiation-induced tumors continue to be a significant side effect of radiotherapy and radiosurgery, and they are frequently identified as recurrent, numerous, and more aggressive. Prolonged follow-up should definitely not be ignored in patients with antecedents of radiotherapy for a primary cerebral tumor or cranial irradiation for an early age malignancy. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
14. Editorial: Childhood leukemias in Latin America: epidemiology, causality, novel predictive profiles and therapeutic strategies.
- Author
-
Núñez-Enríquez, Juan Carlos, Mejía-Aranguré, Juan Manuel, Cruz-Muñoz, Mario Ernesto, and Pelayo, Rosana
- Subjects
SCIENTIFIC knowledge ,ACUTE promyelocytic leukemia ,MYELOID leukemia ,LYMPHOBLASTIC leukemia ,ACUTE leukemia - Abstract
The editorial "Childhood leukemias in Latin America: epidemiology, causality, novel predictive profiles and therapeutic strategies" explores the unique challenges and advancements in understanding and treating childhood leukemias in Latin America. The research topic delves into genetic, environmental, and socio-environmental factors contributing to high incidence rates of childhood leukemia in the region. It also highlights the importance of enhancing diagnosis, treatment, and follow-up strategies through collaborative efforts and technological integration to improve patient outcomes and reduce disparities in care. The authors propose initiatives to expand technological integration, improve risk stratification, strengthen resource distribution, promote preventive actions, foster collaboration, investigate disease determinants, and bring cutting-edge technology to vulnerable regions to enhance pediatric leukemia care in Latin America. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
15. Long-term survival outcome of childhood acute myeloid leukemia: a 43-year experience in Thailand, a resource-limited country.
- Author
-
Sripornsawan, Pornpun, Chavananon, Shevachut, Kittivisuit, Sirinthip, Songthawee, Natsaruth, McNeil, Edward B., and Chotsampancharoen, Thirachit
- Subjects
- *
LEUKOCYTE count , *ACUTE myeloid leukemia , *RESOURCE-limited settings , *CHILD patients , *SURVIVAL rate - Abstract
Although there have been advances in treating pediatric patients with acute myeloid leukemia (AML) in developed countries, outcomes in low- to middle-income countries remain poor. The goal of this study was to investigate the outcomes in children with AML who were treated at a tertiary care center in Thailand. We divided the study into 4 research periods based on the chemotherapy protocols employed. The 5-year probabilities of event-free survival (pEFS) rates for periods 1–4 were 19.0%, 20.6%, 17.4%, and 37.3% (p value = 0.32), while the 5-year probabilities of overall survival (pOS) rates were 19.0%, 24.7%, 18.7%, and 42.5% (p value = 0.18), respectively. The multivariable model indicated an improvement in 5-year pOS between periods 1 and 4 (p value = 0.04). Age, white blood cell count, and study period were significant predictors of survival outcomes. The pOS of AML patients improved over time, increasing from 19.0% to 42.5%. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Hypoalbuminemia in children with acute lymphoblastic leukemia: relation to asparaginase therapy and impact on high dose methotrexate elimination.
- Author
-
Christensen, Sophie Rex, Jensen, Christina Friis, Heldrup, Jesper, Taylor, Zachary, Ramsey, Laura B., and Rosthøj, Steen
- Subjects
- *
SERUM albumin , *LYMPHOBLASTIC leukemia , *ASPARAGINASE , *ACUTE leukemia , *METHOTREXATE , *ALBUMINS - Abstract
Purpose: High-dose methotrexate (HDMTX) therapy is an important component in treatment regimens for acute lymphoblastic leukemia (ALL). Courses are associated with a risk of renal injury, delayed elimination, and increased systemic toxicity. Recently hypoalbuminemia has been recognized as yet another risk factor. Methods: To examine the impact of serum albumin we reviewed 325 HDMTX 5 g/m2 courses in a cohort of 51 children treated on the NOPHO ALL 2008 protocol, dividing the courses into four groups with different levels of baseline albumin (A < 25 g/L, B 25–29 g/L, C 30–34 g/L and D ≥ 35 g/L). Results: Hypoalbuminemia was present in 51% of the courses, mostly in the early phases of chemotherapy while asparaginase therapy is ongoing, and especially if given less than 2 weeks after a dose (78%). Hypoalbuminemia had a significant impact on the end-of-infusion serum MTX, depending on the degree of hypoalbuminemia: MTX > 150 µM was seen in 37%, 32%, 20% and 8% in groups A to D. Serum albumin < 30 g/L was significantly associated with low MTX clearance < 10 L/h/1.73m2 (78% vs. 36%) and high AUC ≥ 1000 µM*h (44% vs. 31%). The frequency of rising creatinine or prolonged elimination was not increased, but the risk of stomatitis was significantly higher (42% vs. 19%). Conclusion: Low serum albumin is caused by concurrent asparaginase therapy and has a clinically significant impact on MTX disposition. Guidelines for administering HDMTX may need adjustment if serum albumin < 30 g/L, and, if possible, HDMTX courses should not be scheduled soon after asparaginase doses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Nonchromosomal birth defects and risk of childhood acute leukemia: An assessment in 15 000 leukemia cases and 46 000 controls from the Childhood Cancer and Leukemia International Consortium
- Author
-
Lupo, Philip J, Chambers, Tiffany M, Mueller, Beth A, Clavel, Jacqueline, Dockerty, John D, Doody, David R, Erdmann, Friederike, Ezzat, Sameera, Filippini, Tommaso, Hansen, Johnni, Heck, Julia E, Infante‐Rivard, Claire, Kang, Alice Y, Magnani, Corrado, Malagoli, Carlotta, Marcotte, Erin L, Metayer, Catherine, Bailey, Helen D, Mora, Ana M, Ntzani, Evangelia, Petridou, Eleni Th, Pombo‐de‐Oliveira, Maria S, Rashed, Wafaa M, Roman, Eve, Schüz, Joachim, Wesseling, Catharina, Spector, Logan G, and Scheurer, Michael E
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Hematology ,Rare Diseases ,Pediatric Cancer ,Cancer ,Pediatric ,Clinical Research ,Childhood Leukemia ,2.1 Biological and endogenous factors ,Child ,Humans ,Infant ,Risk Factors ,Leukemia ,Myeloid ,Acute ,Birth Weight ,Logistic Models ,Case-Control Studies ,Surveys and Questionnaires ,acute lymphoblastic leukemia ,acute myeloid leukemia ,birth defects ,childhood leukemia ,epidemiology ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
Although recent studies have demonstrated associations between nonchromosomal birth defects and several pediatric cancers, less is known about their role on childhood leukemia susceptibility. Using data from the Childhood Cancer and Leukemia International Consortium, we evaluated associations between nonchromosomal birth defects and childhood leukemia. Pooling consortium data from 18 questionnaire-based and three registry-based case-control studies across 13 countries, we used multivariable logistic regression models to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between a spectrum of birth defects and leukemia. Our analyses included acute lymphoblastic leukemia (ALL, n = 13 115) and acute myeloid leukemia (AML, n = 2120) cases, along with 46 172 controls. We used the false discovery rate to account for multiple comparisons. In the questionnaire-based studies, the prevalence of birth defects was 5% among cases vs 4% in controls, whereas, in the registry-based studies, the prevalence was 11% among cases vs 7% in controls. In pooled adjusted analyses, there were several notable associations, including (1) digestive system defects and ALL (OR = 2.70, 95% CI: 1.46-4.98); (2) congenital anomalies of the heart and circulatory system and AML (OR = 2.86, 95% CI: 1.81-4.52) and (3) nervous system defects and AML (OR = 4.23, 95% CI: 1.50-11.89). Effect sizes were generally larger in registry-based studies. Overall, our results could point to novel genetic and environmental factors associated with birth defects that could also increase leukemia susceptibility. Additionally, differences between questionnaire- and registry-based studies point to the importance of complementary sources of birth defect phenotype data when exploring these associations.
- Published
- 2024
18. Malignant A-to-I RNA editing by ADAR1 drives T cell acute lymphoblastic leukemia relapse via attenuating dsRNA sensing
- Author
-
Rivera, Maria, Zhang, Haoran, Pham, Jessica, Isquith, Jane, Zhou, Qingchen Jenny, Balaian, Larisa, Sasik, Roman, Enlund, Sabina, Mark, Adam, Ma, Wenxue, Holm, Frida, Fisch, Kathleen M, Kuo, Dennis John, Jamieson, Catriona, and Jiang, Qingfei
- Subjects
Biological Sciences ,Pediatric Cancer ,Stem Cell Research - Nonembryonic - Human ,Rare Diseases ,Stem Cell Research ,Genetics ,Hematology ,Pediatric ,Cancer ,Childhood Leukemia ,5.1 Pharmaceuticals ,Humans ,Chronic Disease ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,RNA Editing ,RNA ,Double-Stranded ,T-Lymphocytes ,CP: Cancer ,CP: Immunology ,RNA editing ,epitranscriptome ,leukemia-initiating cells ,pediatric leukemia ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Leukemia-initiating cells (LICs) are regarded as the origin of leukemia relapse and therapeutic resistance. Identifying direct stemness determinants that fuel LIC self-renewal is critical for developing targeted approaches. Here, we show that the RNA-editing enzyme ADAR1 is a crucial stemness factor that promotes LIC self-renewal by attenuating aberrant double-stranded RNA (dsRNA) sensing. Elevated adenosine-to-inosine editing is a common attribute of relapsed T cell acute lymphoblastic leukemia (T-ALL) regardless of molecular subtype. Consequently, knockdown of ADAR1 severely inhibits LIC self-renewal capacity and prolongs survival in T-ALL patient-derived xenograft models. Mechanistically, ADAR1 directs hyper-editing of immunogenic dsRNA to avoid detection by the innate immune sensor melanoma differentiation-associated protein 5 (MDA5). Moreover, we uncover that the cell-intrinsic level of MDA5 dictates the dependency on the ADAR1-MDA5 axis in T-ALL. Collectively, our results show that ADAR1 functions as a self-renewal factor that limits the sensing of endogenous dsRNA. Thus, targeting ADAR1 presents an effective therapeutic strategy for eliminating T-ALL LICs.
- Published
- 2024
19. Targeting IGF2BP3 enhances antileukemic effects of menin-MLL inhibition in MLL-AF4 leukemia
- Author
-
Lin, Tasha L, Jaiswal, Amit K, Ritter, Alexander J, Reppas, Jenna, Tran, Tiffany M, Neeb, Zachary T, Katzman, Sol, Thaxton, Michelle L, Cohen, Amanda, Sanford, Jeremy R, and Rao, Dinesh S
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Pediatric ,Cancer ,Orphan Drug ,Hematology ,Rare Diseases ,Pediatric Cancer ,Childhood Leukemia ,Genetics ,2.1 Biological and endogenous factors ,5.1 Pharmaceuticals ,Humans ,Myeloid-Lymphoid Leukemia Protein ,Leukemia ,Transcription Factors ,Cell Differentiation ,Oncogene Proteins ,Fusion ,Cardiovascular medicine and haematology - Abstract
AbstractRNA-binding proteins (RBPs) are emerging as a novel class of therapeutic targets in cancer, including in leukemia, given their important role in posttranscriptional gene regulation, and have the unexplored potential to be combined with existing therapies. The RBP insulin-like growth factor 2 messenger RNA-binding protein 3 (IGF2BP3) has been found to be a critical regulator of MLL-AF4 leukemogenesis and represents a promising therapeutic target. Here, we study the combined effects of targeting IGF2BP3 and menin-MLL interaction in MLL-AF4-driven leukemia in vitro and in vivo, using genetic inhibition with CRISPR-Cas9-mediated deletion of Igf2bp3 and pharmacologic inhibition of the menin-MLL interaction with multiple commercially available inhibitors. Depletion of Igf2bp3 sensitized MLL-AF4 leukemia to the effects of menin-MLL inhibition on cell growth and leukemic initiating cells in vitro. Mechanistically, we found that both Igf2bp3 depletion and menin-MLL inhibition led to increased differentiation in vitro and in vivo, seen in functional readouts and by gene expression analyses. IGF2BP3 knockdown had a greater effect on increasing survival and attenuating disease than pharmacologic menin-MLL inhibition with small molecule MI-503 alone and showed enhanced antileukemic effects in combination. Our work shows that IGF2BP3 is an oncogenic amplifier of MLL-AF4-mediated leukemogenesis and a potent therapeutic target, providing a paradigm for targeting leukemia at both the transcriptional and posttranscriptional level.
- Published
- 2024
20. Pre- and Postnatal Exposures to Tobacco Smoking and Survival of Childhood Acute Lymphoblastic and Myeloid Leukemias in California, United States.
- Author
-
Metayer, Catherine, Morimoto, Libby M, Kang, Alice Y, Sanchez Alvarez, Jacklyn, and Winestone, Lena E
- Subjects
Epidemiology ,Biomedical and Clinical Sciences ,Public Health ,Health Sciences ,Hematology ,Prevention ,Tobacco ,Clinical Research ,Pediatric ,Cancer ,Tobacco Smoke and Health ,Rare Diseases ,Childhood Leukemia ,Pediatric Cancer ,3.1 Primary prevention interventions to modify behaviours or promote wellbeing ,Respiratory ,Good Health and Well Being ,Female ,Pregnancy ,Adult ,Child ,Humans ,Tobacco Smoke Pollution ,Case-Control Studies ,Tobacco Smoking ,Risk Factors ,California ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Leukemia ,Myeloid ,Acute ,Prenatal Exposure Delayed Effects ,Tobacco Products ,Medical and Health Sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundTobacco smoke adversely affects the prognosis of adult cancers including myeloid leukemia, but less is known in children.MethodsWe evaluated whether pre- and postnatal exposures to tobacco smoke decrease 5-year survival of 1,235 childhood acute lymphoblastic leukemia (ALL) and 188 childhood acute myeloid leukemia (AML) cases derived from a population-based case-control study in California. Cases were diagnosed between 1995 and 2015 (median follow-up time of 13.2 years overall). We obtained data on tobacco smoking (before conception, during pregnancy, after birth), parental education and income, clinical features, and vital status through 2020. Cox proportional hazards regression was used to calculate hazard ratios (HR) and 95% confidence intervals (CI) for mortality associated with smoking, adjusting for sociodemographic characteristics and risk group (ALL only).ResultsAbout 23% of mothers and 39% of fathers reported smoking and 130 children with ALL and 52 with AML died within 5 years. For AML, increased risks of death were observed among children whose fathers smoked before conception compared with nonsmoking fathers [HR = 1.41; 95% confidence interval (CI), 0.95-3.44 and 3.47; 95% CI, 1.37-8.81, respectively for
- Published
- 2024
21. Chemotherapy resistance in acute myeloid leukemia is mediated by A20 suppression of spontaneous necroptosis
- Author
-
Culver-Cochran, Ashley E, Hassan, Aishlin, Hueneman, Kathleen, Choi, Kwangmin, Ma, Averil, VanCauwenbergh, Brett, O’Brien, Eric, Wunderlich, Mark, Perentesis, John P, and Starczynowski, Daniel T
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Oncology and Carcinogenesis ,Cancer ,Pediatric Cancer ,Rare Diseases ,Hematology ,Pediatric ,Genetics ,Stem Cell Research ,Childhood Leukemia ,Humans ,Necroptosis ,Tumor Necrosis Factor alpha-Induced Protein 3 ,Leukemia ,Myeloid ,Acute ,Drug Resistance ,Neoplasm ,NF-kappa B ,Receptor-Interacting Protein Serine-Threonine Kinases ,Cell Line ,Tumor ,Anthracyclines ,Cytarabine ,Animals ,Female ,Male ,Mice ,Middle Aged - Abstract
Acute myeloid leukemia (AML) is a deadly hematopoietic malignancy. Although many patients achieve complete remission with standard induction therapy, a combination of cytarabine and anthracycline, ~40% of patients have induction failure. These refractory patients pose a treatment challenge, as they do not respond to salvage therapy or allogeneic stem cell transplant. Herein, we show that AML patients who experience induction failure have elevated expression of the NF-κB target gene tumor necrosis factor alpha-induced protein-3 (TNFAIP3/A20) and impaired necroptotic cell death. A20High AML are resistant to anthracyclines, while A20Low AML are sensitive. Loss of A20 in AML restores sensitivity to anthracycline treatment by inducing necroptosis. Moreover, A20 prevents necroptosis in AML by targeting the necroptosis effector RIPK1, and anthracycline-induced necroptosis is abrogated in A20High AML. These findings suggest that NF-κB-driven A20 overexpression plays a role in failed chemotherapy induction and highlights the potential of targeting an alternative cell death pathway in AML.
- Published
- 2024
22. Increased AID results in mutations at the CRLF2 locus implicated in Latin American ALL health disparities
- Author
-
Rangel, Valeria, Sterrenberg, Jason N, Garawi, Aya, Mezcord, Vyanka, Folkerts, Melissa L, Calderon, Sabrina E, Garcia, Yadhira E, Wang, Jinglong, Soyfer, Eli M, Eng, Oliver S, Valerin, Jennifer B, Tanjasiri, Sora Park, Quintero-Rivera, Fabiola, Seldin, Marcus M, Masri, Selma, Frock, Richard L, Fleischman, Angela G, and Pannunzio, Nicholas R
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Minority Health ,Pediatric Cancer ,Genetics ,Childhood Leukemia ,Health Disparities ,Rare Diseases ,Cancer ,Pediatric ,Prevention ,Human Genome ,Hematology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Humans ,Cytidine Deaminase ,Mutation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Hispanic or Latino ,Receptors ,Cytokine ,DNA Breaks ,Double-Stranded ,B-Lymphocytes ,Health Status Disparities ,Translocation ,Genetic ,Genetic Loci ,Latin America ,Female - Abstract
Activation-induced cytidine deaminase (AID) is a B cell-specific mutator required for antibody diversification. However, it is also implicated in the etiology of several B cell malignancies. Evaluating the AID-induced mutation load in patients at-risk for certain blood cancers is critical in assessing disease severity and treatment options. We have developed a digital PCR (dPCR) assay that allows us to quantify mutations resulting from AID modification or DNA double-strand break (DSB) formation and repair at sites known to be prone to DSBs. Implementation of this assay shows that increased AID levels in immature B cells increase genome instability at loci linked to chromosomal translocation formation. This includes the CRLF2 locus that is often involved in translocations associated with a subtype of acute lymphoblastic leukemia (ALL) that disproportionately affects Hispanics, particularly those with Latin American ancestry. Using dPCR, we characterize the CRLF2 locus in B cell-derived genomic DNA from both Hispanic ALL patients and healthy Hispanic donors and found increased mutations in both, suggesting that vulnerability to DNA damage at CRLF2 may be driving this health disparity. Our ability to detect and quantify these mutations will potentiate future risk identification, early detection of cancers, and reduction of associated cancer health disparities.
- Published
- 2024
23. Structure-guided functional suppression of AML-associated DNMT3A hotspot mutations
- Author
-
Lu, Jiuwei, Guo, Yiran, Yin, Jiekai, Chen, Jianbin, Wang, Yinsheng, Wang, Gang Greg, and Song, Jikui
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Genetics ,Biological Sciences ,Childhood Leukemia ,Hematology ,Cancer ,Human Genome ,Cancer Genomics ,Rare Diseases ,Pediatric ,Pediatric Cancer ,2.1 Biological and endogenous factors ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,DNA Methyltransferase 3A ,Mutation ,Leukemia ,Myeloid ,Acute ,DNA Methylation ,DNA - Abstract
DNA methyltransferases DNMT3A- and DNMT3B-mediated DNA methylation critically regulate epigenomic and transcriptomic patterning during development. The hotspot DNMT3A mutations at the site of Arg822 (R882) promote polymerization, leading to aberrant DNA methylation that may contribute to the pathogenesis of acute myeloid leukemia (AML). However, the molecular basis underlying the mutation-induced functional misregulation of DNMT3A remains unclear. Here, we report the crystal structures of the DNMT3A methyltransferase domain, revealing a molecular basis for its oligomerization behavior distinct to DNMT3B, and the enhanced intermolecular contacts caused by the R882H or R882C mutation. Our biochemical, cellular, and genomic DNA methylation analyses demonstrate that introducing the DNMT3B-converting mutations inhibits the R882H-/R882C-triggered DNMT3A polymerization and enhances substrate access, thereby eliminating the dominant-negative effect of the DNMT3A R882 mutations in cells. Together, this study provides mechanistic insights into DNMT3A R882 mutations-triggered aberrant oligomerization and DNA hypomethylation in AML, with important implications in cancer therapy.
- Published
- 2024
24. Transcriptome free energy can serve as a dynamic patient-specific biomarker in acute myeloid leukemia
- Author
-
Uechi, Lisa, Vasudevan, Swetha, Vilenski, Daniela, Branciamore, Sergio, Frankhouser, David, O’Meally, Denis, Meshinchi, Soheil, Marcucci, Guido, Kuo, Ya-Huei, Rockne, Russell, and Kravchenko-Balasha, Nataly
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Precision Medicine ,Genetics ,Childhood Leukemia ,Hematology ,Cancer ,Human Genome ,Rare Diseases ,Pediatric ,Pediatric Cancer ,Good Health and Well Being ,Adult ,Animals ,Mice ,Humans ,Child ,Transcriptome ,Gene Expression Profiling ,Leukemia ,Myeloid ,Acute ,Biomarkers ,Tumor ,Phenotype ,Bioinformatics and computational biology - Abstract
Acute myeloid leukemia (AML) is prevalent in both adult and pediatric patients. Despite advances in patient categorization, the heterogeneity of AML remains a challenge. Recent studies have explored the use of gene expression data to enhance AML diagnosis and prognosis, however, alternative approaches rooted in physics and chemistry may provide another level of insight into AML transformation. Utilizing publicly available databases, we analyze 884 human and mouse blood and bone marrow samples. We employ a personalized medicine strategy, combining state-transition theory and surprisal analysis, to assess the RNA transcriptome of individual patients. The transcriptome is transformed into physical parameters that represent each sample's steady state and the free energy change (FEC) from that steady state, which is the state with the lowest free energy.We found the transcriptome steady state was invariant across normal and AML samples. FEC, representing active molecular processes, varied significantly between samples and was used to create patient-specific barcodes to characterize the biology of the disease. We discovered that AML samples that were in a transition state had the highest FEC. This disease state may be characterized as the most unstable and hence the most therapeutically targetable since a change in free energy is a thermodynamic requirement for disease progression. We also found that distinct sets of ongoing processes may be at the root of otherwise similar clinical phenotypes, implying that our integrated analysis of transcriptome profiles may facilitate a personalized medicine approach to cure AML and restore a steady state in each patient.
- Published
- 2024
25. Single-cell RNA sequencing of a new transgenic t(8;21) preleukemia mouse model reveals regulatory networks promoting leukemic transformation
- Author
-
Yan, Ming, Liu, Mengdan, Davis, Amanda G, Stoner, Samuel A, and Zhang, Dong-Er
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Rare Diseases ,Childhood Leukemia ,Pediatric Cancer ,Cancer Genomics ,Genetics ,Pediatric ,Cancer ,Human Genome ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Human ,Hematology ,Stem Cell Research - Nonembryonic - Non-Human ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Humans ,Mice ,Animals ,Preleukemia ,RUNX1 Translocation Partner 1 Protein ,Leukemia ,Myeloid ,Acute ,Core Binding Factor Alpha 2 Subunit ,Animals ,Genetically Modified ,Sequence Analysis ,RNA ,Oncogene Proteins ,Fusion ,Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Cardiovascular medicine and haematology ,Clinical sciences ,Oncology and carcinogenesis - Abstract
T(8;21)(q22;q22), which generates the AML1-ETO fusion oncoprotein, is a common chromosomal abnormality in acute myeloid leukemia (AML) patients. Despite having favorable prognosis, 40% of patients will relapse, highlighting the need for innovative models and application of the newest technologies to study t(8;21) leukemogenesis. Currently, available AML1-ETO mouse models have limited utility for studying the pre-leukemic stage because AML1-ETO produces mild hematopoietic phenotypes and no leukemic transformation. Conversely, overexpression of a truncated variant, AML1-ETO9a (AE9a), promotes fully penetrant leukemia and is too potent for studying pre-leukemic changes. To overcome these limitations, we devised a germline-transmitted Rosa26 locus AE9a knock-in mouse model that moderately overexpressed AE9a and developed leukemia with long latency and low penetrance. We observed pre-leukemic alterations in AE9a mice, including skewing of progenitors towards granulocyte/monocyte lineages and replating of stem and progenitor cells. Next, we performed single-cell RNA sequencing to identify specific cell populations that contribute to these pre-leukemic phenotypes. We discovered a subset of common myeloid progenitors that have heightened granulocyte/monocyte bias in AE9a mice. We also observed dysregulation of key hematopoietic transcription factor target gene networks, blocking cellular differentiation. Finally, we identified Sox4 activation as a potential contributor to stem cell self-renewal during the pre-leukemic stage.
- Published
- 2024
26. Hyperemesis gravidarum and the risk of childhood cancer – A case-control study in Denmark
- Author
-
Orimoloye, Helen T, Deng, Chuanjie, Hansen, Johnni, Olsen, Jorn, Saechao, Chai, Ritz, Beate, and Heck, Julia E
- Subjects
Biomedical and Clinical Sciences ,Epidemiology ,Health Services and Systems ,Health Sciences ,Oncology and Carcinogenesis ,Hematology ,Rare Diseases ,Cancer ,Neurosciences ,Prevention ,Neuroblastoma ,Orphan Drug ,Pediatric ,Pediatric Cancer ,Childhood Leukemia ,2.1 Biological and endogenous factors ,Reproductive health and childbirth ,Pregnancy ,Female ,Humans ,Child ,Case-Control Studies ,Hyperemesis Gravidarum ,Mothers ,Denmark ,Hyperemesis gravidarum ,Childhood cancer ,Acute lymphoblastic ,Leukemia ,Non-Hodgkin 's lymphoma ,Antinauseants ,Antiemetics ,Non-Hodgkin's lymphoma ,Public Health and Health Services ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
ObjectiveOnly a few studies have reported on the association between hyperemesis gravidarum and the risk of childhood cancer. We examined possible associations in this population-based study in Denmark.MethodsPediatric cancer cases (n = 6420) were ascertained from the Denmark Cancer Registry among children born between 1977 and 2013. Twenty-five controls were matched to each case by sex and birth date from the Central Person Registry (n = 160500). Mothers with hyperemesis gravidarum were ascertained from the National Patient Register. The risk of childhood cancer was estimated using conditional logistic regression. In a separate analysis, we examined pregnancy prescription of antinauseant medications, ascertained from the National Pharmaceutical Register, to determine associations with childhood cancers.ResultsIn Denmark, hyperemesis gravidarum was associated with an increased risk of childhood cancer [all types combined; Odds Ratio (OR) = 1.43, 95% confidence interval (CI) 1.12, 1.81; n = 73 exposed cases). Hyperemesis gravidarum was also associated with an increased risk of neuroblastoma (OR = 2.52, 95% CI 1.00, 6.36; n = 5 exposed cases), acute lymphoblastic leukemia (OR = 1.63, 95% CI 0.98, 2.72; n = 16 exposed cases), and non-Hodgkin's lymphoma (OR = 2.41, 95% CI 0.95, 6.08; n = 5 exposed cases). We observed no childhood cancer risk increase from antinauseant prescriptions (OR = 1.05, 95% CI 0.84, 1.30; n = 91 exposed cases).ConclusionOur results are suggestive of an association between hyperemesis gravidarum and the overall cancer risk in offspring, particularly for neuroblastoma. Mothers with hyperemesis gravidarum should be closely monitored and receive appropriate treatment during pregnancy.
- Published
- 2023
27. Gene-Environment Analyses Reveal Novel Genetic Candidates with Prenatal Tobacco Exposure in Relation to Risk for Childhood Acute Lymphoblastic Leukemia.
- Author
-
Zhong, Charlie, Li, Shaobo, Arroyo, Katti, Morimoto, Libby M, de Smith, Adam J, Metayer, Catherine, Ma, Xiaomei, Kogan, Scott C, Gauderman, W James, and Wiemels, Joseph L
- Subjects
Epidemiology ,Biomedical and Clinical Sciences ,Public Health ,Health Sciences ,Prevention ,Childhood Leukemia ,Cancer ,Hematology ,Women's Health ,Genetics ,Pediatric Cancer ,Pregnancy ,Pediatric ,Rare Diseases ,Tobacco ,Tobacco Smoke and Health ,Clinical Research ,Perinatal Period - Conditions Originating in Perinatal Period ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Good Health and Well Being ,Infant ,Newborn ,Female ,Humans ,Maternal Exposure ,Smoking ,DNA Methylation ,Transcription Factors ,Tobacco Smoke Pollution ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prenatal Exposure Delayed Effects ,Medical and Health Sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundAssociations between maternal tobacco exposure during pregnancy and childhood acute lymphoblastic leukemia (ALL) have yielded mixed results. This may be due to biases in self-reported smoking or other differences in individual-level risk factors. We utilized a biological marker of maternal tobacco exposure to evaluate the association between maternal tobacco exposure during pregnancy, genetics, and subsequent childhood ALL risk in two large population-based studies of childhood ALL in California.MethodsMaternal exposure to tobacco smoke was assessed with a validated methylation marker (cg05575921) of the aryl hydrocarbon receptor repressor (AHRR) gene in newborn dried blood spots. We adjusted for sex, birthweight, gestational age, mode of delivery, year of birth, AHRR quantitative trait locus (mQTL) rs77111113, and a polygenetic risk score for childhood ALL. We additionally adjusted for principal components in a gene-environment interaction testing method that incorporates gene-only and environment-only effects along with interactions.ResultsAHRR hypomethylation overall was not associated with childhood ALL. In gene-environment interaction testing, several genetic variants displayed significant interaction with AHRR hypomethylation and childhood ALL.ConclusionsOur results suggest that novel candidates in PTPRK and DPP6 may play a role in tobacco-related leukemogenesis. Further research is necessary to better understand the effects of tobacco and these variants on childhood ALL risk.ImpactDespite the lack of an overall "main effect," tobacco exposure during pregnancy affects childhood ALL risk depending on specific genetic variants.
- Published
- 2023
28. Optimizing early phase clinical trial washout periods: a report from the Therapeutic Advances in Childhood Leukemia and Lymphoma consortium.
- Author
-
Schafer, Eric S, Rushing, Teresa, Crews, Kristine R, Annesley, Colleen, Colace, Susan I, Kaiser, Nicole, Pommert, Lauren, Ramsey, Laura B, Sabnis, Himalee S, Wong, Kenneth, Chang, Bill H, Cooper, Todd M, Shah, Nirali N, Rheingold, Susan R, Place, Andrew E, Chi, Yueh-Yun, Bhojwani, Deepa, Wayne, Alan S, and Bernhardt, M Brooke
- Subjects
- *
COMPREHENSION testing , *EMPLOYMENT portfolios , *CONSORTIA , *TASK forces , *LANGUAGE policy - Abstract
Background The National Cancer Institute (NCI) issued a 2021 memorandum adopting the American Society of Clinical Oncology (ASCO) and Friends of Cancer Research (Friends) task force recommendations to broaden clinical study eligibility criteria. They recommended that washout periods be eliminated for most prior cancer therapy and when required to utilize evidence- and/or rationale-based criteria. The Therapeutic Advances in Childhood Leukemia and Lymphoma (TACL) consortium responded to this guidance. Methods A TACL task force reviewed the consortium's research portfolio, the relevant literature and guidance documents from ASCO-Friends, NCI, and US Food and Drug Administration to make expert consensus and evidence-based recommendations for modernizing, broadening, and codifying TACL-study washout periods while ensuring consistency with pediatric ethics, and federal regulations. TACL's screening log was reviewed to estimate the impact that updated washout periods would have on patient inclusivity and recruitment. Results Over a 19-year period, 42 (14.6% of all screened ineligible patients [n = 287]) patients were identified as excluded from TACL early phase studies exclusively because of not meeting washout criteria. An additional 6 (2.1%) did not meet washout and at least 1 other exclusion criterion. A new TACL washout guidance document was developed and then adopted for use. Where washout criteria were not eliminated, rationale- and/or evidenced-based criteria were established with citation. Conclusion In an effort to reduce unnecessary exclusion from clinical trials, TACL created rationale- and/or evidenced-based washout period standards largely following guidance from the NCI and ASCO-Friends recommendations. These new, expanded eligibility criteria are expected to increase access to TACL clinical trials while maintaining safety and scientific excellence. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. A Pilot Study Conducted at a Tertiary Cancer Care Center, Evaluating the Serum Asparaginase Activity in Children Suffering from Acute Lymphoblastic Leukemia after the Administration of Biosimilar Pegaspargase.
- Author
-
Venkatagiri, Archana Melavarige, Bhat, Vasudeva K., Asok, Arjun, and Prabhu, Krishnananda
- Subjects
- *
LYMPHOBLASTIC leukemia , *ACUTE leukemia , *ASPARAGINASE , *GRAM-negative bacteria , *CANCER treatment - Abstract
Introduction L-asparaginase is considered to be the most important component in the treatment of acute lymphoblastic leukemia (ALL). Intensifying the use of Lasparaginase during treatment for ALL has resulted in a significant rise in the percentage of children and adolescents who are cured of the disease. Asparaginase trough activity more than or equal to 100 IU/L on day 7 has been found to be the desired activity level in all childhood leukemia patients. Objectives Due to the paucity of data on biosimilar pegaspargase in the upfront setting, we planned this prospective pilot study to evaluate the levels of serum asparaginase activity (SAA) after biosimilar pegaspargase infusion. Materials and Methods It is a prospective, single-center, pilot study of 10 pediatric ALL patients for the duration of 6 months. All children less than 18 years with ALL on treatment with curative intent and receiving pegaspargase and who provided informed consent were included in this study. The enzymatic spectrophotometric method was used to determine SAA, and it was measured on the 7th and 14th days after the first dosage of pegaspargase-asparaginase, as well as on the 14th day after the second dose of pegaspargase-asparaginase, while toxicity was charted according to Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Results From 10 patients with a median age of 5.5 years, a grand total of 29 samples were taken for analysis. Children who received pegaspargase had either B-ALL or T-ALL. After the first dose, mean-SD (standard deviation), SAA levels at day 7 was 131.3±38 IU/L and at Day 14 was 94.8±8 IU/L. After the second dose, mean±SD SAA level at day 14 was 86.1±15 IU/L. No patient had clinical hypersensitivity reaction and no patient reported any asparaginase-related toxicity. One patient died due to sepsis, infection with multidrug-resistant gram-negative bacteria. Conclusions Biosimilar pegaspargasemaintained good SAA levels 7 and 14 days after infusion. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Fluoroquinolone Prophylaxis in Children With Cancer: A Pro/Con Discussion.
- Author
-
Vasileiadi, Eleana, Lloyd, Kevin M, Fisher, Brian T, and Hanisch, Benjamin
- Subjects
- *
BACTEREMIA prevention , *MEDICAL protocols , *FLUOROQUINOLONES , *DRUG side effects , *INFECTION control , *CANCER patient medical care , *DRUG resistance in microorganisms , *CANCER patients , *FEVER , *DISCUSSION , *PEDIATRICS , *QUINOLONE antibacterial agents , *CANCER chemotherapy , *LYMPHOBLASTIC leukemia , *CHILDREN - Abstract
There are conflicting recommendations on whether to use or not to use fluoroquinolone prophylaxis in pediatric oncology patients. An international pediatric clinical practice guideline (CPG) recommends administering levofloxacin prophylaxis in patients with acute myeloblastic leukemia and relapsed acute lymphoblastic leukemia receiving intensive chemotherapy as this practice has been found to reduce episodes of fever and bacteremia. A separate European CPG does not recommend levofloxacin prophylaxis because of concerns for adverse effects, including potentiation of fluoroquinolone resistance and possible increased resistance to other classes of antibiotics. The nuance of the decision to give or not give prophylaxis is discussed in the context of published evidence defining the risks and benefits of levofloxacin prophylaxis for pediatric leukemia patients at high risk for bacterial infection. Knowledge gaps are also identified to guide further investigations to optimize the use of fluoroquinolone prophylaxis in pediatric patients receiving chemotherapy for cancer or undergoing a hematopoietic cell transplantation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Exploring cell-derived extracellular vesicles in peripheral blood and bone marrow of B-cell acute lymphoblastic leukemia pediatric patients: proof-of-concept study.
- Author
-
Magalhães-Gama, Fábio, Silvestrini, Marina Malheiros Araújo, Ferreira Neves, Juliana Costa, Araújo, Nilberto Dias, Alves-Hanna, Fabíola Silva, Kerr, Marlon Wendell Athaydes, Carvalho, Maria Perpétuo Socorro Sampaio, Tarragô, Andréa Monteiro, Pontes, Gemilson Soares, Martins-Filho, Olindo Assis, Malheiro, Adriana, Teixeira-Carvalho, Andreá, and Costa, Allyson Guimarães
- Subjects
CHILD patients ,EXTRACELLULAR vesicles ,LYMPHOBLASTIC leukemia ,BONE marrow ,T cells - Abstract
Extracellular vesicles (EVs) are heterogeneous, phospholipid membrane enclosed particles that are secreted by healthy and cancerous cells. EVs are present in diverse biological fluids and have been associated with the severity of diseases, which indicates their potential as biomarkers for diagnosis, prognosis and as therapeutic targets. This study investigated the phenotypic characteristics of EVs derived from peripheral blood (PB) and bone marrow (BM) in pediatric patients with B-cell acute lymphoblastic leukemia (B-ALL) during different treatment stages. PB and BM plasma were collected from 20 B-ALL patients at three time points during induction therapy, referred to as: diagnosis baseline (D0), day 15 of induction therapy (D15) and the end of the induction therapy (D35). In addition, PB samples were collected from 10 healthy children at a single time point. The EVs were measured using CytoFLEX S flow cytometer. Calibration beads were employed to ensure accurate size analysis. The following, fluorescent-labeled specific cellular markers were used to label the EVs: Annexin V (phosphatidylserine), CD235a (erythrocyte), CD41a (platelet), CD51 (endothelial cell), CD45 (leukocyte), CD66b (neutrophil), CD14 (monocyte), CD3 (T lymphocyte), CD19, CD34 and CD10 (B lymphoblast/leukemic blast). Our results demonstrate that B-ALL patients had a marked production of EV-CD51/61
+ , EV-CD10+ , EV-CD19+ and EV-CD10+ CD19+ (double-positive) with a decrease in EV-CD41a+ on D0. However, the kinetics and signature of production during induction therapy revealed a clear decline in EV-CD10+ and EV-CD19+ , with an increase of EV-CD41a+ on D35. Furthermore, B-ALL patients showed a complex biological network, exhibiting distinct profiles on D0 and D35. Interestingly, fold change and ROC curve analysis demonstrated that EV-CD10+ CD19+ were associated with B-ALL patients, exhibited excellent clinical performance and standing out as a potential diagnostic biomarker. In conclusion, our data indicate that EVs represent a promising field of investigation in B-ALL, offering the possibility of identifying potential biomarkers and therapeutic targets. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
32. Efficacy of the Allosteric MEK Inhibitor Trametinib in Relapsed and Refractory Juvenile Myelomonocytic Leukemia: A Report from the Children's Oncology Group
- Author
-
Stieglitz, Elliot, Lee, Alex, Angus, Steven P, Davis, Christopher, Barkauskas, Donald, Hall, David, Kogan, Scott C, Meyer, Julia, Rhodes, Steven D, Xuei, Xiaoling, Shannon, Kevin, Loh, Mignon L, Fox, Elizabeth, and Weigel, Brenda J
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Oncology and Carcinogenesis ,Stem Cell Research - Nonembryonic - Non-Human ,Minority Health ,Hematology ,Stem Cell Research ,Genetics ,Rare Diseases ,Pediatric Cancer ,Childhood Leukemia ,Cancer ,Pediatric ,Clinical Trials and Supportive Activities ,Transplantation ,Clinical Research ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,Inflammatory and immune system ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Paediatrics and Reproductive Medicine ,Immunology ,Biochemistry and cell biology ,Cardiovascular medicine and haematology ,Paediatrics - Abstract
Background: Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative neoplasm of infants and toddlers. Upfront therapies typically include high-dose cytarabine or azacitidine, but the only definitive treatment is hematopoietic stem cell transplantation (HSCT). While HSCT cures ~50% of patients, the prognosis is dismal for those who relapse. In the absence of a second HSCT, patients who relapse have a 2-year overall survival of ~10%. JMML is initiated by germline and somatic driver mutations in NF1, KRAS, NRAS, PTPN11, and CBL. These mutations converge on Ras signaling, leading to elevated levels of active Ras-GTP in specific cell lineages. Genetically engineered mouse (GEM) models accurately model key molecular, biologic, and biochemical features of JMML. Preclinical trials of the allosteric MEK inhibitors in Kras and Nf1 mutant mice demonstrated dramatic phenotypic responses with reduction in white blood cell counts, resolution of splenomegaly, and reversion to normal erythropoiesis. Based on the promising efficacy signal in GEM models, we evaluated trametinib, an orally bioavailable allosteric inhibitor of MEK1/2, in a prospective clinical trial in children with relapsed or refractory JMML to determine the overall response rate to trametinib. Results: Ten infants and children with JMML (median age 23.6 months) were enrolled and all were evaluable for safety and efficacy. Patients received age-adjusted dosing of trametinib for 28-day cycles and could remain on study for up to 12 cycles in the absence of disease progression or toxicity. A clonal Ras pathway mutation was confirmed in the blood and/or bone marrow of all patients. The objective response rate was 50% (two complete and three partial clinical responses). Four patients proceeded to HSCT after receiving protocol therapy and remain alive in complete remission with undetectable levels of the Ras pathway mutation identified at enrollment. Three additional patients completed 12 cycles of trametinib and continue to receive off-protocol therapy 6-24 months later with no change in the variant allele frequency of the underlying Ras pathway mutation. The remaining three patients had progressive disease with two demonstrating molecular evolution by the end of cycle 2. Paired pre- and post-trametinib RNASeq and proteomic analyses confirmed on-target biochemical effects of trametinib with down-regulation of both Ras/MAPK pathway related gene expression and MEK1/2 kinase activity, respectively. To gain deeper insight into how trametinib might affect distinct populations of hematopoietic cells, we generated single cell RNASeq data before and after treatment. The most prominent finding was a reduction in the proportions of classical and non-classical monocytes and broad downregulation of immune-related pathways in all cell populations. However, the downregulation of MAPK signaling genes was confined to specific cell types including, macrophages, classical monocytes, and granulocyte-monocyte progenitors, which uniquely displayed downregulated KRAS-related signatures following treatment. High DNA methylation and the presence of >1 mutation at enrollment correlated with lack of response to trametinib. Conclusions: We conducted an open label, phase 2 trial of trametinib in children with relapsed or refractory JMML. This is the first completed study of a MEK inhibitor in any hematologic malignancy in children. The trial met its primary objective and demonstrated a 50% objective response rate with 70% of patients bridging to a successful HSCT or completing the maximum 12 cycles permitted on study. The three patients who completed 12 cycles continue to receive trametinib off study for as long as 2 years. Although limited by a small number of patients, the long-term survival, and correlative molecular analyses from this trial raise provocative questions about whether certain patients with JMML might be spared the long-term adverse health risks of genotoxic HSCT conditioning regimens based on favorable molecular characteristics at diagnosis. This hypothesis will now be tested in a national, risk-stratified therapeutic trial (NCT05849662) of trametinib in combination with azacitidine in newly diagnosed patients. The authors would like to acknowledge funding and support from: NCTN Operations Center Grant (U10CA180886), NCTN Statistics & Data Center Grant (U10CA180899), and the St. Baldrick's Foundation.
- Published
- 2023
33. Allosteric SHP2 inhibition increases apoptotic dependency on BCL2 and synergizes with venetoclax in FLT3- and KIT-mutant AML
- Author
-
Popescu, Bogdan, Stahlhut, Carlos, Tarver, Theodore C, Wishner, Sydney, Lee, Bianca J, Peretz, Cheryl AC, Luck, Cuyler, Phojanakong, Paul, Serrano, Juan Antonio Camara, Hongo, Henry, Rivera, Jose M, Xirenayi, Simayijiang, Chukinas, John A, Steri, Veronica, Tasian, Sarah K, Stieglitz, Elliot, and Smith, Catherine C
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Pediatric Cancer ,Childhood Leukemia ,Rare Diseases ,Cancer ,Pediatric ,Orphan Drug ,Hematology ,5.1 Pharmaceuticals ,Good Health and Well Being ,Humans ,Cell Line ,Tumor ,Apoptosis ,Leukemia ,Myeloid ,Acute ,Protein Kinase Inhibitors ,Proto-Oncogene Proteins c-bcl-2 ,fms-Like Tyrosine Kinase 3 ,FLT3 ,SHP2 ,acute myeloid leukemia ,apoptosis ,cancer ,drug synergy ,targeted therapies ,Biomedical and clinical sciences - Abstract
Mutations in the receptor tyrosine kinases (RTKs) FLT3 and KIT are frequent and associated with poor outcomes in acute myeloid leukemia (AML). Although selective FLT3 inhibitors (FLT3i) are clinically effective, remissions are short-lived due to secondary resistance characterized by acquired mutations constitutively activating the RAS/MAPK pathway. Hereby, we report the pre-clinical efficacy of co-targeting SHP2, a critical node in MAPK signaling, and BCL2 in RTK-driven AML. The allosteric SHP2 inhibitor RMC-4550 suppresses proliferation of AML cell lines with FLT3 and KIT mutations, including cell lines with acquired resistance to FLT3i. We demonstrate that pharmacologic SHP2 inhibition unveils an Achilles' heel of RTK-driven AML, increasing apoptotic dependency on BCL2 via MAPK-dependent mechanisms, including upregulation of BMF and downregulation of MCL1. Consequently, RMC-4550 and venetoclax are synergistically lethal in AML cell lines and in clinically relevant xenograft models. Our results provide mechanistic rationale and pre-clinical evidence for co-targeting SHP2 and BCL2 in RTK-driven AML.
- Published
- 2023
34. Framework humanization optimizes potency of anti-CD72 nanobody CAR-T cells for B-cell malignancies
- Author
-
Temple, William C, Nix, Matthew A, Naik, Akul, Izgutdina, Adila, Huang, Benjamin J, Wicaksono, Gianina, Phojanakong, Paul, Serrano, Juan Antonio Camara, Young, Elizabeth P, Ramos, Emilio, Salangsang, Fernando, Steri, Veronica, Xirenayi, Simayijiang, Hermiston, Michelle, Logan, Aaron C, Stieglitz, Elliot, and Wiita, Arun P
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Childhood Leukemia ,Biotechnology ,Pediatric Cancer ,Orphan Drug ,Hematology ,Pediatric ,Cancer ,Gene Therapy ,Genetics ,Immunotherapy ,Lymphoma ,Lymphatic Research ,Immunization ,Rare Diseases ,5.1 Pharmaceuticals ,5.2 Cellular and gene therapies ,Animals ,Humans ,Receptors ,Chimeric Antigen ,Immunotherapy ,Adoptive ,T-Lymphocytes ,Camelids ,New World ,Burkitt Lymphoma ,Recurrence ,Antigens ,Differentiation ,B-Lymphocyte ,Antigens ,CD ,Cell Engineering ,Hematologic Neoplasms ,Translational Medical Research ,Oncology and carcinogenesis - Abstract
BackgroundApproximately 50% of patients who receive anti-CD19 CAR-T cells relapse, and new immunotherapeutic targets are urgently needed. We recently described CD72 as a promising target in B-cell malignancies and developed nanobody-based CAR-T cells (nanoCARs) against it. This cellular therapy design is understudied compared with scFv-based CAR-T cells, but has recently become of significant interest given the first regulatory approval of a nanoCAR in multiple myeloma.MethodsWe humanized our previous nanobody framework regions, derived from llama, to generate a series of humanized anti-CD72 nanobodies. These nanobody binders were inserted into second-generation CD72 CAR-T cells and were evaluated against preclinical models of B cell acute lymphoblastic leukemia and B cell non-Hodgkin's lymphoma in vitro and in vivo. Humanized CD72 nanoCARs were compared with parental ("NbD4") CD72 nanoCARs and the clinically approved CD19-directed CAR-T construct tisangenlecleucel. RNA-sequencing, flow cytometry, and cytokine secretion profiling were used to determine differences between the different CAR constructs. We then used affinity maturation on the parental NbD4 construct to generate high affinity binders against CD72 to test if higher affinity to CD72 improved antitumor potency.ResultsToward clinical translation, here we humanize our previous nanobody framework regions, derived from llama, and surprisingly discover a clone ("H24") with enhanced potency against B-cell tumors, including patient-derived samples after CD19 CAR-T relapse. Potentially underpinning improved potency, H24 has moderately higher binding affinity to CD72 compared with a fully llama framework. However, further affinity maturation (KD
- Published
- 2023
35. Structural surfaceomics reveals an AML-specific conformation of integrin β2 as a CAR T cellular therapy target
- Author
-
Mandal, Kamal, Wicaksono, Gianina, Yu, Clinton, Adams, Jarrett J, Hoopmann, Michael R, Temple, William C, Izgutdina, Adila, Escobar, Bonell Patiño, Gorelik, Maryna, Ihling, Christian H, Nix, Matthew A, Naik, Akul, Xie, William H, Hübner, Juwita, Rollins, Lisa A, Reid, Sandy M, Ramos, Emilio, Kasap, Corynn, Steri, Veronica, Serrano, Juan Antonio Camara, Salangsang, Fernando, Phojanakong, Paul, McMillan, Melanie, Gavallos, Victor, Leavitt, Andrew D, Logan, Aaron C, Rooney, Cliona M, Eyquem, Justin, Sinz, Andrea, Huang, Benjamin J, Stieglitz, Elliot, Smith, Catherine C, Moritz, Robert L, Sidhu, Sachdev S, Huang, Lan, and Wiita, Arun P
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Cancer ,Hematology ,Orphan Drug ,Gene Therapy ,Rare Diseases ,Pediatric Cancer ,Pediatric ,Childhood Leukemia ,Genetics ,Immunotherapy ,Biotechnology ,5.2 Cellular and gene therapies ,5.1 Pharmaceuticals ,Humans ,Receptors ,Chimeric Antigen ,T-Lymphocytes ,Integrins ,Immunotherapy ,Adoptive ,Leukemia ,Myeloid ,Acute ,Oncology and carcinogenesis - Abstract
Safely expanding indications for cellular therapies has been challenging given a lack of highly cancer-specific surface markers. Here we explore the hypothesis that tumor cells express cancer-specific surface protein conformations that are invisible to standard target discovery pipelines evaluating gene or protein expression, and these conformations can be identified and immunotherapeutically targeted. We term this strategy integrating cross-linking mass spectrometry with glycoprotein surface capture 'structural surfaceomics'. As a proof of principle, we apply this technology to acute myeloid leukemia (AML), a hematologic malignancy with dismal outcomes and no known optimal immunotherapy target. We identify the activated conformation of integrin β2 as a structurally defined, widely expressed AML-specific target. We develop and characterize recombinant antibodies to this protein conformation and show that chimeric antigen receptor T cells eliminate AML cells and patient-derived xenografts without notable toxicity toward normal hematopoietic cells. Our findings validate an AML conformation-specific target antigen and demonstrate a tool kit for applying these strategies more broadly.
- Published
- 2023
36. A phase 1b study of venetoclax and alvocidib in patients with relapsed/refractory acute myeloid leukemia
- Author
-
Jonas, Brian A, Hou, Jing‐Zhou, Roboz, Gail J, Alvares, Caroline L, Jeyakumar, Deepa, Edwards, John R, Erba, Harry P, Kelly, Richard J, Röllig, Christoph, Fiedler, Walter, Brackman, Deanna, Siddani, Satya R, Chyla, Brenda, Hilger‐Rolfe, Jacqueline, and Watts, Justin M
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Hematology ,Clinical Research ,Clinical Trials and Supportive Activities ,Orphan Drug ,Childhood Leukemia ,Patient Safety ,Pediatric Cancer ,Pediatric ,Rare Diseases ,Cancer ,6.1 Pharmaceuticals ,6.2 Cellular and gene therapies ,Good Health and Well Being ,Humans ,Antineoplastic Combined Chemotherapy Protocols ,Bridged Bicyclo Compounds ,Heterocyclic ,Leukemia ,Myeloid ,Acute ,acute myeloid leukemia ,alvocidib ,BCL-2 ,drug resistance ,MCL-1 ,venetoclax ,Cardiorespiratory Medicine and Haematology ,Immunology ,Cardiovascular medicine and haematology - Abstract
Relapsed/refractory (R/R) Acute Myeloid Leukemia (AML) is a genetically complex and heterogeneous disease with a poor prognosis and limited treatment options. Thus, there is an urgent need to develop therapeutic combinations to overcome drug resistance in AML. This open-label, multicenter, international, phase 1b study evaluated the safety, efficacy, and pharmacokinetics of venetoclax in combination with alvocidib in patients with R/R AML. Patients were treated with escalating doses of venetoclax (400, 600, and 800 mg QD, orally, days 1-28) and alvocidib (45 and 60 mg/m2 , intravenously, days 1-3) in 28-day cycles. The combination was found to be safe and tolerable, with no maximum tolerated dose reached. Drug-related Grade ≥3 adverse events were reported in 23 (65.7%) for venetoclax and 24 (68.6%) for alvocidib. No drug-related AEs were fatal. Gastrointestinal toxicities, including diarrhea, nausea, and vomiting were notable and frequent; otherwise, the toxicities reported were consistent with the safety profile of both agents. The response rate was modest (complete remission [CR] + incomplete CR [CRi], 11.4%; CR + CRi + partial response rate + morphologic leukemia-free state, 20%). There was no change in alvocidib pharmacokinetics with increasing doses of venetoclax. However, when venetoclax was administered with alvocidib, AUC24 and Cmax decreased by 18% and 19%, respectively. A recommended phase 2 dose was not established due to lack of meaningful increase in efficacy across all cohorts compared to what was previously observed with each agent alone. Future studies could consider the role of the sequence, dosing, and the use of a more selective MCL1 inhibitor for the R/R AML population.
- Published
- 2023
37. Evaluating genomic polygenic risk scores for childhood acute lymphoblastic leukemia in Latinos
- Author
-
Jeon, Soyoung, Lo, Ying Chu, Morimoto, Libby M, Metayer, Catherine, Ma, Xiaomei, Wiemels, Joseph L, de Smith, Adam J, and Chiang, Charleston WK
- Subjects
Biological Sciences ,Genetics ,Childhood Leukemia ,Human Genome ,Pediatric Cancer ,Prevention ,Pediatric ,Hematology ,Cancer ,Rare Diseases ,Biotechnology ,Cancer Genomics ,Child ,Humans ,Genetic Predisposition to Disease ,Genetic Risk Score ,Genome-Wide Association Study ,Genomics ,Hispanic or Latino ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,United States ,White ,Racial Groups ,acute lymphoblastic leukemia ,latinos ,polygenic risk scores ,risk prediction - Abstract
The utility of polygenic risk score (PRS) models has not been comprehensively evaluated for childhood acute lymphoblastic leukemia (ALL), the most common type of cancer in children. Previous PRS models for ALL were based on significant loci observed in genome-wide association studies (GWASs), even though genomic PRS models have been shown to improve prediction performance for a number of complex diseases. In the United States, Latino (LAT) children have the highest risk of ALL, but the transferability of PRS models to LAT children has not been studied. In this study, we constructed and evaluated genomic PRS models based on either non-Latino White (NLW) GWAS or a multi-ancestry GWAS. We found that the best PRS models performed similarly between held-out NLW and LAT samples (PseudoR2 = 0.086 ± 0.023 in NLW vs. 0.060 ± 0.020 in LAT), and can be improved for LAT if we performed GWAS in LAT-only (PseudoR2 = 0.116 ± 0.026) or multi-ancestry samples (PseudoR2 = 0.131 ± 0.025). However, the best genomic models currently do not have better prediction accuracy than a conventional model using all known ALL-associated loci in the literature (PseudoR2 = 0.166 ± 0.025), which includes loci from GWAS populations that we could not access to train genomic PRS models. Our results suggest that larger and more inclusive GWASs may be needed for genomic PRS to be useful for ALL. Moreover, the comparable performance between populations may suggest a more oligogenic architecture for ALL, where some large effect loci may be shared between populations. Future PRS models that move away from the infinite causal loci assumption may further improve PRS for ALL.
- Published
- 2023
38. A Novel bispecific T-cell engager (BiTE) targeting CD22 and CD3 has both in vitro and in vivo activity and synergizes with blinatumomab in an acute lymphoblastic leukemia (ALL) tumor model
- Author
-
Meckler, Joshua F, Levis, Daniel J, Vang, Daniel P, and Tuscano, Joseph M
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Lymphoma ,Cancer ,Pediatric Cancer ,Genetics ,Vaccine Related ,Childhood Leukemia ,Orphan Drug ,Pediatric ,Rare Diseases ,Immunotherapy ,Lymphatic Research ,Biotechnology ,Hematology ,Immunization ,5.1 Pharmaceuticals ,5.2 Cellular and gene therapies ,Humans ,Animals ,Mice ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Antineoplastic Agents ,T-Lymphocytes ,Antibodies ,Bispecific ,Antigens ,CD19 ,Acute lymphoblastic leukemia ,Bispecific T-cell engager ,CD22 ,CD3 ,Oncology and carcinogenesis - Abstract
Immunotherapy has revolutionized cancer therapy. Two recently FDA-approved immunotherapies for B-cell malignancies target CD19, in the form of a Bispecific T-Cell Engager (BiTE) antibody construct or chimeric antigen receptor T (CAR-T) cells. Blinatumomab, an FDA-approved BiTE, binds to CD19 on B cells and to CD3 on T cells, mediating effector-target cell contact and T-cell activation that results in effective elimination of target B cells. Although CD19 is expressed by essentially all B-cell malignancies at clinical presentation, relapses with loss or reduction in CD19 surface expression are increasingly recognized as a cause of treatment failure. Therefore, there is a clear need to develop therapeutics for alternate targets. We have developed a novel BiTE consisting of humanized anti-CD22 and anti-CD3 single chain variable fragments. Target binding of the anti-CD22 and anti-CD3 moieties was confirmed by flow cytometry. CD22-BiTE promoted in vitro cell-mediated cytotoxicity in a dose and effector: target (E:T)-dependent fashion. Additionally, in an established acute lymphoblastic leukemia (ALL) xenograft mouse model, CD22-BiTE demonstrated tumor growth inhibition, comparable to blinatumomab. Further, the combination of blinatumomab and CD22-BiTE yielded increased efficacy in vivo when compared to the single agents. In conclusion, we report here the development of a new BiTE with cytotoxic activity against CD22+ cells which could represent an alternate or complementary therapeutic option for B-cell malignancies.
- Published
- 2023
39. A gut instinct for childhood leukemia prevention: microbiome-targeting recommendations aimed at parents and caregivers
- Author
-
Ersen Kameri, Vera Helena Jepsen, Pawel Stachura, Nadine Rüchel, Rigveda Bhave, Leticia Benitez, Fatima Crispi, Eduard Gratacos, Nico Dragano, Stefan Janssen, Arndt Borkhardt, Aleksandra Pandyra, Gesine Kögler, and Ute Fischer
- Subjects
childhood leukemia ,risk factors ,gut microbiome ,prevention ,recommendations ,public health ,Public aspects of medicine ,RA1-1270 - Abstract
Childhood leukemia accounts for 30% of all pediatric cancer cases with acute lymphoblastic leukemia (ALL) being the most common subtype. Involvement of the gut microbiome in ALL development has recently garnered interest due to an increasing recognition of the key contribution the microbiome plays in maintaining the immune system's homeostatic balance. Commensal gut microbiota provide a first line of defense against different pathogens and gut microbiome immaturity has been implicated in ALL pathogenesis. Several environmental factors such as nutrition, mode of delivery, breastfeeding and, early social or livestock contacts are known to alter the composition of the gut microbiota. Variations in these factors influence the risk of childhood leukemia onset. This review aims to elucidate the risk factors influencing microbial composition in the context of childhood ALL. The link between gut microbiome diversity and childhood ALL offers the opportunity to develop risk-reducing strategies that can be communicated to a broad target population of (future) parents and caregivers for childhood leukemia prevention. Here, we summarize evidence on how promoting a diverse gut microbiome in newborns through simple measures such as increasing social contacts early in life may decrease the risk of developing ALL in these children later on.
- Published
- 2025
- Full Text
- View/download PDF
40. Entospletinib with decitabine in acute myeloid leukemia with mutant TP53 or complex karyotype: A phase 2 substudy of the Beat AML Master Trial
- Author
-
Duong, Vu H, Ruppert, Amy S, Mims, Alice S, Borate, Uma, Stein, Eytan M, Baer, Maria R, Stock, Wendy, Kovacsovics, Tibor, Blum, William, Arellano, Martha L, Schiller, Gary J, Olin, Rebecca L, Foran, James M, Litzow, Mark R, Lin, Tara L, Patel, Prapti A, Foster, Matthew C, Redner, Robert L, Al‐Mansour, Zeina, Cogle, Christopher R, Swords, Ronan T, Collins, Robert H, Vergilio, Jo‐Anne, Heerema, Nyla A, Rosenberg, Leonard, Yocum, Ashley O, Marcus, Sonja, Chen, Timothy, Druggan, Franchesca, Stefanos, Mona, Gana, Theophilus J, Shoben, Abigail B, Druker, Brian J, Burd, Amy, Byrd, John C, Levine, Ross L, and Boyiadzis, Michael M
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Childhood Leukemia ,Pediatric ,Rare Diseases ,Hematology ,Cancer ,Clinical Research ,Orphan Drug ,Aging ,Clinical Trials and Supportive Activities ,Pediatric Cancer ,6.1 Pharmaceuticals ,Humans ,Decitabine ,Tumor Suppressor Protein p53 ,Leukemia ,Myeloid ,Acute ,Karyotype ,Treatment Outcome ,Antineoplastic Combined Chemotherapy Protocols ,acute myeloid leukemia ,decitabine ,entospletinib ,hypomethylating agents ,tumor protein p53 ,Public Health and Health Services ,Oncology & Carcinogenesis ,Oncology and carcinogenesis ,Public health - Abstract
BackgroundPatients with acute myeloid leukemia (AML) who have tumor protein p53 (TP53) mutations or a complex karyotype have a poor prognosis, and hypomethylating agents are often used. The authors evaluated the efficacy of entospletinib, an oral inhibitor of spleen tyrosine kinase, combined with decitabine in this patient population.MethodsThis was a multicenter, open-label, phase 2 substudy of the Beat AML Master Trial (ClinicalTrials.gov identifier NCT03013998) using a Simon two-stage design. Eligible patients aged 60 years or older who had newly diagnosed AML with mutations in TP53 with or without a complex karyotype (cohort A; n = 45) or had a complex karyotype without TP53 mutation (cohort B; n = 13) received entospletinib 400 mg twice daily with decitabine 20 mg/m2 on days 1-10 every 28 days for up to three induction cycles, followed by up to 11 consolidation cycles, in which decitabine was reduced to days 1-5. Entospletinib maintenance was given for up to 2 years. The primary end point was complete remission (CR) and CR with hematologic improvement by up to six cycles of therapy.ResultsThe composite CR rates for cohorts A and B were 13.3% (95% confidence interval, 5.1%-26.8%) and 30.8% (95% confidence interval, 9.1%-61.4%), respectively. The median duration of response was 7.6 and 8.2 months, respectively, and the median overall survival was 6.5 and 11.5 months, respectively. The study was stopped because the futility boundary was crossed in both cohorts.ConclusionsThe combination of entospletinib and decitabine demonstrated activity and was acceptably tolerated in this patient population; however, the CR rates were low, and overall survival was short. Novel treatment strategies for older patients with TP53 mutations and complex karyotype remain an urgent need.
- Published
- 2023
41. Autoimmune HLA Alleles and Neoepitope Presentation Predict Post-Allogenic Transplant Relapse
- Author
-
Castro, Andrea, Goodman, Aaron M, Rane, Zachary, Talwar, James V, Frampton, Garrett M, Morris, Gerald P, Lippman, Scott M, Zhang, Xinlian, Kurzrock, Razelle, and Carter, Hannah
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Oncology and Carcinogenesis ,Immunology ,Hematology ,Childhood Leukemia ,Genetics ,Minority Health ,Stem Cell Research ,Clinical Research ,Pediatric Cancer ,Pediatric ,Cancer ,Transplantation ,Stem Cell Research - Nonembryonic - Human ,Rare Diseases ,2.1 Biological and endogenous factors ,Good Health and Well Being ,allo-HSCT ,HLA genotype ,neoantigen ,relapse ,acute myeloid leukemia - Abstract
IntroductionAllogeneic hematopoietic stem cell transplantation (allo-HSCT) can cure patients with high-risk myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). However, many patients relapse or develop debilitating graft-versus-host disease. Transplant restores T-cell reactivity against tumor cells, implicating patient human leukocyte antigen (HLA)-dependent antigen presentation via the major histocompatibility complex as a determinant of response. We sought to identify characteristics of the HLA genotype that influence response in allo-HSCT patients.MethodsWe collected HLA genotype and panel-based somatic mutation profiles for 55 patients with AML and MDS and available data treated at the University of California San Diego Moores Cancer Center between May 2012 and January 2019. We evaluated characteristics of the HLA genotype relative to relapse-free time and overall survival (OS) post-allo-HSCT using univariable and multivariable regression.ResultsIn multivariable regression, the presence of an autoimmune allele was significantly associated with relapse-free time (hazard ratio [HR], 0.25; p = 0.01) and OS (HR, 0.16; p < 0.005). The better potential of the donor HLA type to present peptides harboring driver mutations trended toward better relapse-free survival (HR, 0.45; p = 0.07) and significantly correlated with longer OS (HR, 0.33; p = 0.01) though only a minority of cases had an HLA mismatch.ConclusionIn this single institution retrospective study of patients receiving allo-HSCT for relapsed AML/MDS, characteristics of an individual's HLA genotype (presence of an autoimmune allele and potential of the donor HLA to better present peptides representing driver mutations) were significantly associated with better outcomes. These findings suggest that HLA type may guide the optimal application of allo-HSCT and merit evaluation in larger cohorts. ClinicalTrials.gov Identifier: NCT02478931.
- Published
- 2023
42. Transcriptomic classes of BCR-ABL1 lymphoblastic leukemia.
- Author
-
Kim, Jaeseung C, Chan-Seng-Yue, Michelle, Ge, Sabrina, Zeng, Andy GX, Ng, Karen, Gan, Olga I, Garcia-Prat, Laura, Flores-Figueroa, Eugenia, Woo, Tristan, Zhang, Amy Xin Wei, Arruda, Andrea, Chithambaram, Shivapriya, Dobson, Stephanie M, Khoo, Amanda, Khan, Shahbaz, Ibrahimova, Narmin, George, Ann, Tierens, Anne, Hitzler, Johann, Kislinger, Thomas, Dick, John E, McPherson, John D, Minden, Mark D, and Notta, Faiyaz
- Subjects
Humans ,Fusion Proteins ,bcr-abl ,Protein Kinase Inhibitors ,Gene Expression Profiling ,Cell Differentiation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Transcriptome ,Hematology ,Pediatric Cancer ,Rare Diseases ,Human Genome ,Pediatric Research Initiative ,Stem Cell Research ,Cancer ,Childhood Leukemia ,Pediatric ,Genetics ,2.1 Biological and endogenous factors ,Aetiology ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
In BCR-ABL1 lymphoblastic leukemia, treatment heterogeneity to tyrosine kinase inhibitors (TKIs), especially in the absence of kinase domain mutations in BCR-ABL1, is poorly understood. Through deep molecular profiling, we uncovered three transcriptomic subtypes of BCR-ABL1 lymphoblastic leukemia, each representing a maturation arrest at a stage of B-cell progenitor differentiation. An earlier arrest was associated with lineage promiscuity, treatment refractoriness and poor patient outcomes. A later arrest was associated with lineage fidelity, durable leukemia remissions and improved patient outcomes. Each maturation arrest was marked by specific genomic events that control different transition points in B-cell development. Interestingly, these events were absent in BCR-ABL1+ preleukemic stem cells isolated from patients regardless of subtype, which supports that transcriptomic phenotypes are determined downstream of the leukemia-initialing event. Overall, our data indicate that treatment response and TKI efficacy are unexpected outcomes of the differentiation stage at which this leukemia transforms.
- Published
- 2023
43. Artificial intelligence reveals the predictions of hematological indexes in children with acute leukemia
- Author
-
Zhangkai J. Cheng, Haiyang Li, Mingtao Liu, Xing Fu, Li Liu, Zhiman Liang, Hui Gan, and Baoqing Sun
- Subjects
Childhood leukemia ,Cancer early screening ,Machine learning ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Childhood leukemia is a prevalent form of pediatric cancer, with acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) being the primary manifestations. Timely treatment has significantly enhanced survival rates for children with acute leukemia. This study aimed to develop an early and comprehensive predictor for hematologic malignancies in children by analyzing nutritional biomarkers, key leukemia indicators, and granulocytes in their blood. Using a machine learning algorithm and ten indices, the blood samples of 826 children with ALL and 255 children with AML were compared to a control group of 200 healthy children. The study revealed notable differences, including higher indicators in boys compared to girls and significant variations in most biochemical indicators between leukemia patients and healthy children. Employing a random forest model resulted in an area under the curve (AUC) of 0.950 for predicting leukemia subtypes and an AUC of 0.909 for forecasting AML. This research introduces an efficient diagnostic tool for early screening of childhood blood cancers and underscores the potential of artificial intelligence in modern healthcare.
- Published
- 2024
- Full Text
- View/download PDF
44. Results of salvage therapy with mini-hyper-CVD and inotuzumab ozogamicin with or without blinatumomab in pre-B acute lymphoblastic leukemia
- Author
-
Kantarjian, Hagop, Haddad, Fadi G, Jain, Nitin, Sasaki, Koji, Short, Nicholas J, Loghavi, Sanam, Kanagal-Shamanna, Rashmi, Jorgensen, Jeffrey, Khouri, Issa, Kebriaei, Partow, Alvarado, Yesid, Kadia, Tapan, Paul, Shilpa, Garcia-Manero, Guillermo, Dabaja, Bouthaina, Yilmaz, Musa, Jacob, Jovitta, Garris, Rebecca, O’Brien, Susan, Ravandi, Farhad, and Jabbour, Elias
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Oncology and Carcinogenesis ,Clinical Research ,Rare Diseases ,Stem Cell Research - Nonembryonic - Human ,Pediatric Cancer ,Stem Cell Research ,Hematology ,Childhood Leukemia ,Cancer ,Pediatric ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,Good Health and Well Being ,Adult ,Humans ,Antibodies ,Bispecific ,Cardiovascular Diseases ,Inotuzumab Ozogamicin ,Methotrexate ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Salvage Therapy ,Blinatumomab ,Chemo-immunotherapy ,Inotuzumab ,Outcome ,Philadelphia-negative ALL ,Salvage ,Cardiorespiratory Medicine and Haematology ,Cardiovascular medicine and haematology ,Oncology and carcinogenesis - Abstract
BackgroundHistorically, adults with relapsed-refractory acute lymphoblastic leukemia (ALL) experienced poor outcomes with intensive chemotherapy. This mature analysis explores the benefit of the addition of sequential blinatumomab to low-intensity mini-Hyper-CVD chemotherapy with inotuzumab ozogamicin in this setting.MethodsMini-Hyper-CVD (cyclophosphamide and dexamethasone at 50% dose reduction, no anthracycline, methotrexate at 75% dose reduction, cytarabine at 83% dose reduction) was combined with inotuzumab during the first 4 courses. From Patient #68 and onwards, inotuzumab was given in reduced and fractionated doses, and blinatumomab was added sequentially for 4 courses. Maintenance therapy with prednisone, vincristine, 6-mercaptopurine and methotrexate was given for 12 courses, and blinatumomab for 4 additional courses.ResultsAmong 110 patients (median age, 37 years) treated, 91 (83%) responded (complete response, 69 patients, 63%). Measurable residual disease negativity was documented in 75 patients (82% of responders). Fifty-three patients (48%) received allogeneic stem cell transplantation (SCT). Hepatic sinusoidal obstruction syndrome occurred in 9/67 patients (13%) on the original inotuzumab schedule and in 1/43 (2%) on the modified schedule. With a median follow-up of 48 months, the median overall survival (OS) was 17 months, and the 3 year OS was 40%. The 3 year OS was 34% with mini-Hyper-CVD plus inotuzumab and 52% with additional blinatumomab (P = 0.16). By landmark analysis at 4 months, the 3 year OS was 54%, similar between patients who did or did not receive allogeneic SCT.ConclusionLow-intensity mini-Hyper-CVD plus inotuzumab with or without blinatumomab showed efficacy in patients with relapsed-refractory ALL, with better survival after the addition of blinatumomab. Trial registration The trial was registered on clinicaltrials.gov with the identifier NCT01371630.
- Published
- 2023
45. The American Society of Pediatric Hematology Oncology workforce, productivity, and fellowship assessment: Current state of the workforce
- Author
-
Hastings, Caroline, Borinstein, Scott C, Bergsagel, D John, Hagstrom, J Nathan, Hooker, Ryan, Nugent, Diane J, Hudspeth, Michelle, and Workgroup, the American Society of Pediatric Hematology Oncology Workforce Publication
- Subjects
Paediatrics ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Childhood Leukemia ,Pediatric Research Initiative ,Pediatric ,Rare Diseases ,Pediatric Cancer ,Hematology ,Cancer ,Child ,Humans ,United States ,Fellowships and Scholarships ,Education ,Medical ,Graduate ,Medical Oncology ,Workforce ,ASPHO ,fellowship ,workforce ,American Society of Pediatric Hematology Oncology Workforce Publication Workgroup ,Clinical Sciences ,Paediatrics and Reproductive Medicine ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
The American Society of Pediatric Hematology Oncology conducted follow-up workforce surveys in 2017 and 2021 as well as a Pediatric Hematology Oncology Fellowship Program Directors Survey in 2020 to provide an updated review of the current workforce. We provide a comprehensive review and analysis of these results with the goal to provide better understanding of the current landscape in pediatric hematology oncology.
- Published
- 2023
46. Dasatinib/prednisone induction followed by blinatumomab/dasatinib in Ph+ acute lymphoblastic leukemia.
- Author
-
Advani, Anjali S, Moseley, Anna, O'Dwyer, Kristen M, Wood, Brent L, Park, Jae, Wieduwilt, Matthew, Jeyakumar, Deepa, Yaghmour, George, Atallah, Ehab L, Gerds, Aaron T, O'Brien, Susan M, Liesveld, Jane L, Othus, Megan, Litzow, Mark, Stone, Richard M, Sharon, Elad, and Erba, Harry P
- Subjects
Humans ,Prednisone ,Treatment Outcome ,Aged ,Aged ,80 and over ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Dasatinib ,Hematology ,Cancer ,Rare Diseases ,Vaccine Related ,Childhood Leukemia ,Orphan Drug ,Clinical Research ,Pediatric Cancer ,Pediatric ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals - Abstract
Novel treatment strategies are needed for the treatment of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) in older patients. This trial evaluated the feasibility and outcomes with the anti-CD19 bispecific T-cell-engaging antibody, blinatumomab, in combination with dasatinib and steroids. Patients 65 years of age or older with Ph+ or Ph-like ALL (with dasatinib-sensitive fusions/mutations) were eligible and could be newly diagnosed or relapsed/refractory. Induction therapy consisted of dasatinib/prednisone. Patients not achieving response by day 56 proceeded to blinatumomab reinduction therapy. Patients achieving response with induction or reinduction therapy proceeded to blinatumomab/dasatinib postremission therapy for 3 cycles followed by dasatinib/prednisone maintenance. All patients received central nervous system prophylaxis with intrathecal methotrexate for a total of 8 doses. Response was assessed at days 28, 56, and 84 and at additional time points based on response parameters. Measurable residual disease was assessed centrally by 8-color flow cytometry at day 28. A total of 24 eligible patients with newly diagnosed Ph+ ALL were enrolled with a median age of 73 years (range, 65-87 years). This combination was safe and feasible. With a median of 2.7 years of follow-up, 3-year overall survival and disease-free survival were 87% (95% confidence interval [CI], 64-96) and 77% (95% CI, 54-90), respectively. Although longer follow-up is needed, these results are encouraging, and future trials are building on this backbone regimen. This trial was registered at www.clinicaltrials.gov as #NCT02143414.
- Published
- 2023
47. Associations between early‐life and in utero infections and cytomegalovirus‐positive acute lymphoblastic leukemia in children
- Author
-
Gallant, Rachel E, Arroyo, Katti, Metayer, Catherine, Kang, Alice Y, de Smith, Adam J, and Wiemels, Joseph L
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Cancer ,Pediatric Cancer ,Perinatal Period - Conditions Originating in Perinatal Period ,Rare Diseases ,Hematology ,Childhood Leukemia ,Conditions Affecting the Embryonic and Fetal Periods ,Infectious Diseases ,Prevention ,Pediatric ,Clinical Research ,2.1 Biological and endogenous factors ,4.1 Discovery and preclinical testing of markers and technologies ,Infection ,Female ,Pregnancy ,Child ,Humans ,Cytomegalovirus ,Cytomegalovirus Infections ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Polymerase Chain Reaction ,Logistic Models ,childhood infection ,cytomegalovirus ,leukemia etiology ,maternal infection ,pediatric acute lymphoblastic leukemia ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
Childhood infections and cytomegalovirus (CMV) are associated with pediatric acute lymphoblastic leukemia (ALL). CMV dysregulates the host immune system and alters the immune response to subsequent antigenic exposures. We suspect that this immune dysregulation contributes to increased numbers of symptomatic infections in childhood allowing for expansion of pre-leukemic clones. We explored the association between childhood infections, maternal infections during pregnancy and CMV-positive ALL. Using a droplet digital PCR assay, we screened diagnostic ALL bone marrow samples from the California Childhood Leukemia Study (1995-2015) for the presence of CMV DNA identifying CMV-positive and CMV-negative cases. We performed a case-only analysis (n = 524) comparing the number and types of childhood infections and maternal infections during pregnancy between CMV-positive and CMV-negative ALL cases using logistic regression. With increasing numbers of infections in the first 12 months of life, children were more likely to classify to the highest tertile of CMV DNA in the bone marrow at diagnosis (OR: 1.04, 95% CI: 1.01-1.08). Specifically, those reporting cough or flu in the first 12 months were more likely to be CMV-positive at ALL diagnosis (OR: 2.15, 95% CI: 1.06-4.37 and OR: 2.06, 95% CI: 1.17-3.63 respectively). Furthermore, those with a history of maternal infection during pregnancy were more likely to be CMV-positive (OR: 2.12, 95% CI: 1.24-3.62). We hypothesize that children with underlying immune dysregulation develop more symptomatic infections in childhood and ultimately CMV-positive ALL; this underlying immune dysregulation may be due to early immune system alterations via CMV exposure (in utero or early infancy) proposing a potential link between CMV and ALL etiology.
- Published
- 2023
48. Detection and targeting of splicing deregulation in pediatric acute myeloid leukemia stem cells
- Author
-
van der Werf, Inge, Mondala, Phoebe K, Steel, S Kathleen, Balaian, Larisa, Ladel, Luisa, Mason, Cayla N, Diep, Raymond H, Pham, Jessica, Cloos, Jacqueline, Kaspers, Gertjan JL, Chan, Warren C, Mark, Adam, La Clair, James J, Wentworth, Peggy, Fisch, Kathleen M, Crews, Leslie A, Whisenant, Thomas C, Burkart, Michael D, Donohoe, Mary E, and Jamieson, Catriona HM
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Stem Cell Research ,Hematology ,Genetics ,Orphan Drug ,Stem Cell Research - Nonembryonic - Human ,Childhood Leukemia ,Pediatric Cancer ,Regenerative Medicine ,Pediatric ,Human Genome ,Cancer Genomics ,Rare Diseases ,Cancer ,Stem Cell Research - Nonembryonic - Non-Human ,Adult ,Child ,Humans ,Stem Cells ,RNA Splicing ,Leukemia ,Myeloid ,Acute ,Protein Isoforms ,Mutation ,RNA Splicing Factors ,Repressor Proteins ,CD47 ,RBFOX2 ,SF3B1 ,embryonic stem cells ,hematopoietic stem cells ,pediatric AML ,pre-mRNA splicing ,splicing modulation ,Biomedical and clinical sciences - Abstract
Pediatric acute myeloid leukemia (pAML) is typified by high relapse rates and a relative paucity of somatic DNA mutations. Although seminal studies show that splicing factor mutations and mis-splicing fuel therapy-resistant leukemia stem cell (LSC) generation in adults, splicing deregulation has not been extensively studied in pAML. Herein, we describe single-cell proteogenomics analyses, transcriptome-wide analyses of FACS-purified hematopoietic stem and progenitor cells followed by differential splicing analyses, dual-fluorescence lentiviral splicing reporter assays, and the potential of a selective splicing modulator, Rebecsinib, in pAML. Using these methods, we discover transcriptomic splicing deregulation typified by differential exon usage. In addition, we discover downregulation of splicing regulator RBFOX2 and CD47 splice isoform upregulation. Importantly, splicing deregulation in pAML induces a therapeutic vulnerability to Rebecsinib in survival, self-renewal, and lentiviral splicing reporter assays. Taken together, the detection and targeting of splicing deregulation represent a potentially clinically tractable strategy for pAML therapy.
- Published
- 2023
49. Cmpd10357 to treat B-cell acute lymphoblastic leukemia
- Author
-
Lee, Alex Q, Konishi, Hiroaki, Helmke, Elizabeth, Ijiri, Masami, Lerot, Jan Michael A, Hicks, Emma, Chien, Jeremy R, Gorin, Fredric A, and Satake, Noriko
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Childhood Leukemia ,Orphan Drug ,Cancer ,Pediatric Cancer ,Biotechnology ,Hematology ,Pediatric ,Rare Diseases ,Women's Health ,5.1 Pharmaceuticals ,Humans ,Mice ,Animals ,Child ,Apoptosis ,Antineoplastic Agents ,Caspases ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Burkitt Lymphoma ,Cell Line ,Tumor ,Cardiorespiratory Medicine and Haematology ,Immunology ,Cardiovascular medicine and haematology - Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is the most common type of cancer found in children. Although the overall survival rates are now >80%, 15%-20% of pediatric patients relapse, with survival rates subsequently dropping to 5%-10%. Cmpd10357, 3-amino-5-arylamino-6-chloro-N- (diaminomethylene) pyrazine-2-carboximide, is a highly potent, cell-permeant compound recently shown to have cytotoxic effects on solid tumors, including human breast cancer and high-grade gliomas, independent of their proliferative status. Cmpd10357 demonstrated concentration-dependent cytotoxicity in two human B-ALL cell lines, JM1 and Reh, at half-maximal inhibitory concentrations (IC50) of 3.2 and 3.3 μM, respectively. Cmpd10357, at a dose of 5 mg/kg, significantly prolonged survival in our B-ALL xenograft mouse model, with a median survival time of 49.0 days compared with 45.5 days in the control group (p < 0.05). The cytotoxicity of Cmpd10357 demonstrated caspase-independent, nonapoptotic cancer cell demise associated with the nuclear translocation of apoptosis-inducing factor (AIF). The cytotoxicity of Cmpd10357 in B-ALL cells was inhibited by Necrostatin-1 but not by Necrosulfonamide. These studies suggest that an AIF-mediated, caspase-independent necrosis mechanism of Cmpd10357 in B-ALL could be used in combination with traditional apoptotic chemotherapeutic agents.
- Published
- 2023
50. Artificial intelligence reveals the predictions of hematological indexes in children with acute leukemia.
- Author
-
Cheng, Zhangkai J., Li, Haiyang, Liu, Mingtao, Fu, Xing, Liu, Li, Liang, Zhiman, Gan, Hui, and Sun, Baoqing
- Subjects
MACHINE learning ,LYMPHOBLASTIC leukemia ,HEMATOLOGIC malignancies ,ACUTE leukemia ,CHILDHOOD cancer - Abstract
Childhood leukemia is a prevalent form of pediatric cancer, with acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) being the primary manifestations. Timely treatment has significantly enhanced survival rates for children with acute leukemia. This study aimed to develop an early and comprehensive predictor for hematologic malignancies in children by analyzing nutritional biomarkers, key leukemia indicators, and granulocytes in their blood. Using a machine learning algorithm and ten indices, the blood samples of 826 children with ALL and 255 children with AML were compared to a control group of 200 healthy children. The study revealed notable differences, including higher indicators in boys compared to girls and significant variations in most biochemical indicators between leukemia patients and healthy children. Employing a random forest model resulted in an area under the curve (AUC) of 0.950 for predicting leukemia subtypes and an AUC of 0.909 for forecasting AML. This research introduces an efficient diagnostic tool for early screening of childhood blood cancers and underscores the potential of artificial intelligence in modern healthcare. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.