Try a new search

Format these results:

Searched for:

in-biosketch:true

person:carrow01

Total Results:

330


Children's Oncology Group AALL0434: A Phase III Randomized Clinical Trial Testing Nelarabine in Newly Diagnosed T-Cell Acute Lymphoblastic Leukemia

Dunsmore, Kimberly P; Winter, Stuart S; Devidas, Meenakshi; Wood, Brent L; Esiashvili, Natia; Chen, Zhiguo; Eisenberg, Nancy; Briegel, Nikki; Hayashi, Robert J; Gastier-Foster, Julie M; Carroll, Andrew J; Heerema, Nyla A; Asselin, Barbara L; Rabin, Karen R; Zweidler-Mckay, Patrick A; Raetz, Elizabeth A; Loh, Mignon L; Schultz, Kirk R; Winick, Naomi J; Carroll, William L; Hunger, Stephen P
PURPOSE/OBJECTIVE:Nelarabine is effective in inducing remission in patients with relapsed and refractory T-cell acute lymphoblastic leukemia (T-ALL) but has not been fully evaluated in those with newly diagnosed disease. PATIENTS AND METHODS/METHODS:From 2007 to 2014, Children's Oncology Group trial AALL0434 (ClinicalTrials.gov identifier: NCT00408005) enrolled 1,562 evaluable patients with T-ALL age 1-31 years who received the augmented Berlin-Frankfurt-Muenster (ABFM) regimen with a 2 × 2 pseudo-factorial randomization to receive escalating-dose methotrexate (MTX) without leucovorin rescue plus pegaspargase (C-MTX) or high-dose MTX (HDMTX) with leucovorin rescue. Intermediate- and high-risk patients were also randomly assigned after induction to receive or not receive six 5-day courses of nelarabine that was incorporated into ABFM. Patients who experienced induction failure were nonrandomly assigned to HDMTX plus nelarabine. Patients with overt CNS disease (CNS3; ≥ 5 WBCs/μL with blasts) received HDMTX and were randomly assigned to receive or not receive nelarabine. All patients, except those with low-risk disease, received cranial irradiation. RESULTS:= .0001). Toxicities, including neurotoxicity, were acceptable and similar between all four arms. CONCLUSION/CONCLUSIONS:The addition of nelarabine to ABFM therapy improved DFS for children and young adults with newly diagnosed T-ALL without increased toxicity.
PMID: 32813610
ISSN: 1527-7755
CID: 4566952

The NSD2 p.E1099K Mutation is Enriched at Relapse and Confers Drug Resistance in a Cell Context Dependent Manner in Pediatric Acute Lymphoblastic Leukemia

Pierro, Joanna; Saliba, Jason; Narang, Sonali; Sethia, Gunjan; Saint Fleur-Lominy, Shella; Chowdhury, Ashfiyah; Qualls, Anita; Fay, Hannah; Kilberg, Harrison L; Moriyama, Takaya; Fuller, Tori J; Teachey, David T; Schmiegelow, Kjeld; Yang, Jun J; Loh, Mignon L; Brown, Patrick A; Zhang, Jinghui; Ma, Xiaotu; Tsirigos, Aristotelis; Evensen, Nikki A; Carroll, William L
The NSD2 p.E1099K (EK) mutation is observed in 10% of acute lymphoblastic leukemia (ALL) samples with enrichment at relapse indicating a role in clonal evolution and drug resistance. To discover mechanisms that mediate clonal expansion, we engineered B-ALL cell lines (Reh, 697) to overexpress wildtype (WT) and EK NSD2, but observed no differences in proliferation, clonal growth, or chemosensitivity. To address whether NSD2 EK acts collaboratively with other pathways, we used shRNAs to knockdown expression of NSD2 in B-ALL cell lines heterozygous for NSD2 EK (RS4;11, RCH-ACV, SEM). Knockdown resulted in decreased proliferation in all lines, decreased clonal growth in RCH-ACV, and increased sensitivity to cytotoxic chemotherapeutic agents, although the pattern of drug sensitivity varied among cell lines implying that the oncogenic properties of NSD2 mutations are likely cell context specific and rely on cooperative pathways. Knockdown of both Type II and REIIBP EK isoforms had a greater impact than knockdown of Type II alone, suggesting that both SET containing EK isoforms contribute to phenotypic changes driving relapse. Furthermore, in vivo models using both cell lines and patient samples revealed dramatically enhanced proliferation of NSD2 EK compared to WT and reduced sensitivity to 6-mercaptopurine in the relapse sample relative to diagnosis. Finally, EK-mediated changes in chromatin state and transcriptional output differed dramatically among cell lines further supporting a cell context specific role of NSD2 EK. These results demonstrate a unique role of NSD2 EK in mediating clonal fitness through pleiotropic mechanisms dependent on the genetic and epigenetic landscape. Implications: NSD2 p.E1099K mutation leads to drug resistance and a clonal advantage in childhood B-ALL.
PMID: 32332049
ISSN: 1557-3125
CID: 4402512

Integrin α6 mediates drug resistance of acute lymphoblastic B-cell leukemia

Gang, Eun Ji; Kim, Hye Na; Hsieh, Yao Te; Ruan, Yongsheng; Ogana, Heather; Pham, Jennifer; Lee, Solomon; Geng, Huimin; Park, Eugene; Klemm, Lars; Willman, Cheryl L; Carroll, William L; Mittelman, Steven D; Orgel, Etan; Oberley, Matthew James; Parekh, Chintan; Abdel-Azim, Hisham; Bhojwani, Deepa; Wayne, Alan S; De Arcangelis, Adele; Georges-Labouesse, Elisabeth; Wayner, Elizabeth; Bönig, Halvard B; Minasyan, Aspram; Ten Hoeve, Johanna; Graeber, Thomas; Müschen, Markus; Heisterkamp, Nora; Kim, Yong-Mi
Resistance to multimodal chemotherapy continues to limit the prognosis of acute lymphoblastic leukemia (ALL). This occurs in part through a process called adhesion-mediated drug resistance, which depends on ALL cell adhesion to the stroma through adhesion molecules, including integrins. Integrin α6 has been implicated in minimal residual disease in ALL and in the migration of ALL cells to the central nervous system. However, it has not been evaluated in the context of chemotherapeutic resistance. Here, we show that the anti-human α6-blocking antibody P5G10 induces apoptosis in primary ALL in vitro and sensitizes primary ALL cells to chemotherapy or tyrosine kinase inhibition in vitro and in vivo. We further analyzed the underlying mechanism of α6-associated apoptosis using a conditional knockout model of α6 in murine BCR-ABL1+ B-ALL cells and showed that α6-deficient ALL cells underwent apoptosis. In vivo deletion of α6 in combination with tyrosine kinase inhibitor (TKI) treatment was more effective in eradicating ALL than treatment with a TKI (nilotinib) alone. Proteomic analysis revealed that α6 deletion in murine ALL was associated with changes in Src signaling, including the upregulation of phosphorylated Lyn (pTyr507) and Fyn (pTyr530). Thus, our data support α6 as a novel therapeutic target for ALL.
PMID: 32219444
ISSN: 1528-0020
CID: 4358692

Successful Outcomes of Newly Diagnosed T Lymphoblastic Lymphoma: Results From Children's Oncology Group AALL0434

Hayashi, Robert J; Winter, Stuart S; Dunsmore, Kimberly P; Devidas, Meenakshi; Chen, Zhiguo; Wood, Brent L; Hermiston, Michelle L; Teachey, David T; Perkins, Sherrie L; Miles, Rodney R; Raetz, Elizabeth A; Loh, Mignon L; Winick, Naomi J; Carroll, William L; Hunger, Stephen P; Lim, Megan S; Gross, Thomas G; Bollard, Catherine M
PURPOSE/OBJECTIVE:The Children's Oncology Group (COG) protocol AALL0434 evaluated the safety and efficacy of multi-agent chemotherapy with Capizzi-based methotrexate/pegaspargase (C-MTX) in patients with newly diagnosed pediatric T-cell lymphoblastic lymphoma (T-LL) and gained preliminary data using nelarabine in high-risk patients. PATIENTS AND METHODS/METHODS:The trial enrolled 299 patients, age 1-31 years. High-risk (HR) patients had ≥ 1% minimal detectable disease (MDD) in the bone marrow at diagnosis or received prior steroid treatment. Induction failure was defined as failure to achieve a partial response (PR) by the end of the 4-week induction. All patients received the augmented Berlin-Frankfurt-Muenster (ABFM) C-MTX regimen. HR patients were randomly assigned to receive or not receive 6 5-day courses of nelarabine incorporated into ABFM. Patients with induction failure were nonrandomly assigned to ABFM C-MTX plus nelarabine. No patients received prophylactic cranial radiation; however, patients with CNS3 disease (CSF WBC ≥ 5/μL with blasts or cranial nerve palsies, brain/eye involvement, or hypothalamic syndrome) were ineligible. RESULTS:= .55). Disease stage, tumor response, and MDD at diagnosis did not demonstrate thresholds that resulted in differences in EFS. Nelarabine did not show an advantage for HR patients. CNS relapse occurred in only 4 patients. CONCLUSION/CONCLUSIONS:COG AALL0434 produced excellent outcomes in one of the largest trials ever conducted for patients with newly diagnosed T-LL. The COG ABFM regimen with C-MTX provided excellent EFS and OS without cranial radiation.
PMID: 32552472
ISSN: 1527-7755
CID: 4484972

Extensive Remodeling of the Immune Microenvironment in B Cell Acute Lymphoblastic Leukemia

Witkowski, Matthew T; Dolgalev, Igor; Evensen, Nikki A; Ma, Chao; Chambers, Tiffany; Roberts, Kathryn G; Sreeram, Sheetal; Dai, Yuling; Tikhonova, Anastasia N; Lasry, Audrey; Qu, Chunxu; Pei, Deqing; Cheng, Cheng; Robbins, Gabriel A; Pierro, Joanna; Selvaraj, Shanmugapriya; Mezzano, Valeria; Daves, Marla; Lupo, Philip J; Scheurer, Michael E; Loomis, Cynthia A; Mullighan, Charles G; Chen, Weiqiang; Rabin, Karen R; Tsirigos, Aristotelis; Carroll, William L; Aifantis, Iannis
A subset of B cell acute lymphoblastic leukemia (B-ALL) patients will relapse and succumb to therapy-resistant disease. The bone marrow microenvironment may support B-ALL progression and treatment evasion. Utilizing single-cell approaches, we demonstrate B-ALL bone marrow immune microenvironment remodeling upon disease initiation and subsequent re-emergence during conventional chemotherapy. We uncover a role for non-classical monocytes in B-ALL survival, and demonstrate monocyte abundance at B-ALL diagnosis is predictive of pediatric and adult B-ALL patient survival. We show that human B-ALL blasts alter a vascularized microenvironment promoting monocytic differentiation, while depleting leukemia-associated monocytes in B-ALL animal models prolongs disease remission in vivo. Our profiling of the B-ALL immune microenvironment identifies extrinsic regulators of B-ALL survival supporting new immune-based therapeutic approaches for high-risk B-ALL treatment.
PMID: 32470390
ISSN: 1878-3686
CID: 4452012

Impact of Intrathecal Triple Therapy Versus Intrathecal Methotrexate on Disease-Free Survival for High-Risk B-Lymphoblastic Leukemia: Children's Oncology Group Study AALL1131

Salzer, Wanda L; Burke, Michael J; Devidas, Meenakshi; Dai, Yunfeng; Hardy, Kristina K; Kairalla, John A; Gore, Lia; Hilden, Joanne M; Larsen, Eric; Rabin, Karen R; Zweidler-McKay, Patrick A; Borowitz, Michael J; Wood, Brent; Heerema, Nyla A; Carroll, Andrew J; Winick, Naomi; Carroll, William L; Raetz, Elizabeth A; Loh, Mignon L; Hunger, Stephen P
PURPOSE/OBJECTIVE:The high-risk stratum of Children's Oncology Group Study AALL1131 was designed to test the hypothesis that postinduction CNS prophylaxis with intrathecal triple therapy (ITT) including methotrexate, hydrocortisone, and cytarabine would improve the postinduction 5-year disease-free survival (DFS) compared with intrathecal methotrexate (IT MTX), when given on a modified augmented Berlin-Frankfurt-Münster backbone. PATIENTS AND METHODS/METHODS:Children with newly diagnosed National Cancer Institute (NCI) high-risk B-cell acute lymphoblastic leukemia (HR B-ALL) or NCI standard-risk B-ALL with defined minimal residual disease thresholds during induction were randomly assigned to receive postinduction IT MTX or ITT. Patients with CNS3-status disease were not eligible. Postinduction IT therapy was given for a total of 21 to 26 doses. Neurocognitive assessments were performed during therapy and during 1 year off therapy. RESULTS:= .77), respectively. There were no differences in the cumulative incidence of isolated bone marrow relapse, isolated CNS relapse, or combined bone marrow and CNS relapse rates, or in toxicities observed for patients receiving IT MTX compared with ITT. There were no significant differences in neurocognitive outcomes for patients receiving IT MTX compared with ITT. CONCLUSION/CONCLUSIONS:Postinduction CNS prophylaxis with ITT did not improve 5-year DFS for children with HR B-ALL. The standard of care for CNS prophylaxis for children with B-ALL and no overt CNS involvement remains IT MTX.
PMID: 32496902
ISSN: 1527-7755
CID: 4469302

Role of setd2 mutations in the progression and chemoresistance of pediatric lymphoblastic leukemia [Meeting Abstract]

Robbins, G; Chowdhury, A; Greenberg, J; Kilberg, H; Kramer, L; Evensen, N; Carroll, W
Background: Outcomes for children with relapsed B-ALL remain poor, in part due to genetic and epigenetic lesions that confer drug resistance to one or more classes of agents used in therapy. SETD2, an epigenetic modifier, commonly harbors loss of function mutations in relapsed pediatric B-ALL. Prior studies have demonstrated the tumor suppressor function of SETD2 in an AML model as well as its role in resistance to DNA damaging agents, but the role of SETD2 mutations in relapsed B-ALL is not yet understood.
Objective(s): Determine the role of relapse-specific SETD2 loss-offunction mutations in disease progression by measuring proliferation and drug sensitivity in isogenic B-ALL cell lines that recapitulate these mutations. Design/Method: A panel of isogenic B-ALL cell lines (697 and KOPN-8) were generated with knockout of SETD2 using the CRISPR/Cas9 system. Conversely, B-ALL cell lines already harboring SETD2 heterozygous mutations (REH and RCH) had SETD2 expression restored using an inducible vector system. Western blot analysis was used to confirm the relative expression of SETD2 and downstream markers of DNA damage response in engineered cell lines. Isogenic cell lines were plated with or without HEK 293 stromal cells and then exposed to vincristine, etoposide, cytarabine, prednisone, or mercaptopurine for 72 hours. Cell viability of cells plated without stroma was measured using CellTiter-Glo. Apoptosis of cells plated with stroma was measured using flow cytometric analysis of apoptosis markers Annexin V and 7AAD. Relative proliferation of all untreated cell lines were measured over 168 hours using an automated cell counter.
Result(s): 697 and KOPN-8 clones with either a SETD2 heterozygous mutation or compound heterozygous mutations exhibited similar rates of proliferation compared to their respective isogenic controls. The half-maximal inhibitory concentrations (IC50) of all chemotherapy agents were similar in mutant clones and their isogenic controls, regardless of the presence of stromal cells. REH and RCH with reexpression of SETD2 also had similar rates of proliferation and IC50 compared to their isogenic controls. No increase in DNA damage was observed upon knockout of SETD2.
Conclusion(s): Loss of SETD2 expression in B-ALL cell lines does not confer increased resistance to conventional chemotherapy agents, either in isolation orwhen grown in a stromal microenvironment. Conversely, re-expression of SETD2 in lines that already harbored SETD2 mutations does not restore chemosensitivity. We postulate that SETD2 deletions alone may not confer a clonal advantage, but may operate in collaboration with other genetic alterations in a cell context-specific manner
EMBASE:634270106
ISSN: 1545-5017
CID: 4805662

Feasibility of monitoring peripheral blood to detect emerging clones in children with acute lymphoblastic leukemia†

Saliba, Jason; Evensen, Nikki A; Meyer, Julia A; Newman, Daniel; Nersting, Jacob; Narang, Sonali; Ma, Xiaotu; Schmiegelow, Kjeld; Carroll, William L
Relapse-enriched somatic variants drive drug resistance in childhood acute lymphoblastic leukemia. We used digital droplet-based polymerase chain reaction to establish whether relapse-enriched mutations in emerging subclones could be detected in peripheral blood samples before frank relapse. Although limitations in sensitivity for some probes hindered detection of certain variants, we successfully detected variants in NT5C2 and PRPS1 at a fractional abundance of 0.005% to 0.3%, 41 to 116 days before relapse. As mutations in both these genes confer resistance to thiopurines, early detection protocols using peripheral blood could be implemented to preemptively alter maintenance therapy to extinguish resistant clones before overt relapse.
PMID: 32391957
ISSN: 1545-5017
CID: 4437962

Impact of Asparaginase Discontinuation on Outcome in Childhood Acute Lymphoblastic Leukemia: A Report From the Children's Oncology Group

Gupta, Sumit; Wang, Cindy; Raetz, Elizabeth A; Schore, Reuven; Salzer, Wanda L; Larsen, Eric C; Maloney, Kelly W; Mattano, Len A; Carroll, William L; Winick, Naomi J; Hunger, Stephen P; Loh, Mignon L; Devidas, Meenakshi
PURPOSE/OBJECTIVE:substitution or complete ASNase discontinuation is unknown. METHODS:but receiving all doses versus not receiving all ASNase doses. RESULTS:= .03). CONCLUSION/CONCLUSIONS:shortages.
PMID: 32275469
ISSN: 1527-7755
CID: 4377762

Randomized assessment of delayed intensification and two methods for parenteral methotrexate delivery in childhood B-ALL: Children's Oncology Group Studies P9904 and P9905

Winick, Naomi; Martin, Paul L; Devidas, Meenakshi; Shuster, Jonathan; Borowitz, Michael J; Paul Bowman, W; Larsen, Eric; Pullen, Jeanette; Carroll, Andrew; Willman, Cheryl; Hunger, Stephen P; Carroll, William L; Camitta, Bruce M
The delayed intensification (DI) enhanced outcome for patients with acute lymphoblastic leukemia (ALL) treated on BFM 76/79 and CCG 105 after a prednisone-based induction. Childrens Oncology Group protocols P9904/9905 evaluated DI via a post-induction randomization for eligible National Cancer Institute (NCI) standard (SR) and high-risk (HR) patients. A second randomization compared intravenous methotrexate (IV MTX) as a 24- (1 g/m2) vs. 4-h (2 g/m2) infusion. NCI SR patients received a dexamethasone-based three-drug and NCI HR/CNS 3 SR patients a prednisone-based four-drug induction. End induction MRD (minimal residual disease) was obtained but did not impact treatment. DI improved the 10-year continuous complete remission (CCR) rate; 75.5 ± 2.5% vs. 81.8 ± 2.2% p = 0.002, whereas MTX administration did not; 4-h 80.8 ± 1.9%; 24-h 81.4 ± 1.9% (p = 0.7780). Overall survival (OS) at 10 years did not differ with DI: 91.4 ± 1.6% vs. 90.9 ± 1.7% (p = 0.25) without but was higher with the 24-h MTX infusion; 4-h 91.1 ± 1.4%; 24-h 93.9 ± 1.2% (p = 0.0209). MRD predicted outcome; 10-year CCR 87.7 ± 2.2 and 82.1 ± 2.5% when MRD was <0.01% with/without DI (p = 0.007) and 54.3 ± 8% and 44 ± 8% for patients with MRD ≥ 0.01% with/without DI (p = 0.11). DI improved CCR for patients with B-ALL with and without end induction MRD.
PMID: 31728054
ISSN: 1476-5551
CID: 4187032