Searched for: in-biosketch:true
person:carrow01
Outstanding Outcome for Children with Standard Risk-Low (SR-Low) Acute Lymphoblastic Leukemia (ALL) and No Benefit to Intensified Peg-Asparaginase (PEG-ASNase) Therapy: Results of Children's Oncology Group (COG) Study AALL0331 [Meeting Abstract]
Mattano, Leonard A; Devidas, Meenakshi; Friedmann, Alison M; Raetz, Elizabeth A; Loh, Mignon; Buckley, Patrick; Borowitz, Michael J; Carroll, Andrew J; Gastier-Foster, Julie; Heerema, Nyla A; Kadan-Lottick, Nina; Matloub, Yousif; Marshall, David T; Stork, Linda C; Wood, Brent L; Winick, Naomi J; Hunger, Stephen; Carroll, William L; Maloney, Kelly
ISI:000349243505162
ISSN: 1528-0020
CID: 1497722
Pharmacokinetic and Pharmacodynamic Properties of Calaspargase Pegol Escherichia coli L-Asparaginase in the Treatment of Patients With Acute Lymphoblastic Leukemia: Results From Children's Oncology Group Study AALL07P4
Angiolillo, Anne L; Schore, Reuven J; Devidas, Meenakshi; Borowitz, Michael J; Carroll, Andrew J; Gastier-Foster, Julie M; Heerema, Nyla A; Keilani, Taha; Lane, Ashley R; Loh, Mignon L; Reaman, Gregory H; Adamson, Peter C; Wood, Brent; Wood, Charlotte; Zheng, Hao W; Raetz, Elizabeth A; Winick, Naomi J; Carroll, William L; Hunger, Stephen P
PURPOSE: Asparaginase is a critical agent used to treat acute lymphoblastic leukemia (ALL). Pegaspargase (SS-PEG), a pegylated form of Escherichia coli L-asparaginase with a succinimidyl succinate (SS) linker, is the first-line asparaginase product used in Children's Oncology Group (COG) ALL trials. Calaspargase pegol (SC-PEG) replaces the SS linker in SS-PEG with a succinimidyl carbamate linker, creating a more stable molecule. COG AALL07P4 was designed to determine the pharmacokinetic and pharmacodynamic comparability of SC-PEG to SS-PEG in patients with newly diagnosed high-risk (HR) B-cell ALL. PATIENTS AND METHODS: A total of 165 evaluable patients were randomly assigned at a 2:1 ratio to receive SC-PEG at 2,100 (SC-PEG2100; n =69) or 2,500 IU/m2 (SC-PEG2500; n=42) versus SS-PEG 2,500 IU/m2 (SS-PEG2500; n =54) as part of an otherwise identical chemotherapy regimen. The groups were similar demographically, except more female patients received SC-PEG2500. RESULTS: The mean half-life of plasma asparaginase activity for both SC-PEG doses was approximately 2.5x longer than that of SS-PEG2500. The total systemic exposure, as defined by induction area under the curve from time 0 to 25 days, was greater with SC-PEG2500 than with SS-PEG2500 or SC-PEG2100. The proportion of patients with plasma asparaginase activity >/= 100 mIU/mL and >/= 400 mIU/mL was higher in patients who received SC-PEG as compared with SS-PEG2500. After one dose of pegylated asparaginase on induction day 4, plasma asparagine was undetectable for 11 days for SS-PEG2500 and 18 days for both SC-PEG groups. CONCLUSION: SC-PEG2500 achieves a significantly longer period of asparaginase activity above defined thresholds and asparagine depletion compared with SS-PEG2500 and has a comparable toxicity profile in children with HR B-cell ALL.
PMCID:4239306
PMID: 25348002
ISSN: 0732-183x
CID: 1322652
Ikaros deletions in BCR-ABL-negative childhood acute lymphoblastic leukemia are associated with a distinct gene expression signature but do not result in intrinsic chemoresistance
Vitanza, Nicholas A; Zaky, Wafik; Blum, Roy; Meyer, Julia A; Wang, Jinhua; Bhatla, Teena; Morrison, Debra J; Raetz, Elizabeth A; Carroll, William L
BACKGROUND: Ikaros, the product of IKZF1, is a regulator of lymphoid development and polymorphisms in the gene have been associated with the acute lymphoblastic leukemia (ALL). Additionally, IKZF1 deletions and mutations identify high-risk biological subsets of childhood ALL [Georgopoulos et al. Cell 1995;83(2):289-299; Mullighan et al. N Engl J Md 2009;360(5):470-480]. PROCEDURES: To discover the underlying pathways modulated by Ikaros we performed gene expression and gene ontology analysis in IKZF1 deleted primary B-ALL pediatric patient samples. To validate downstream targets we performed qPCR on individual patient samples. We also created IKZF1 knockdown B-ALL cell lines with over 50% reduction of Ikaros, mimicking haplosufficient Ikaros deletions, and again performed qPCR to investigate the downstream targets. Finally, to understand the association of Ikaros deletion with a poor prognosis we challenged our IKZF1 knockdown cell lines with chemotherapy and compared responses to IKZF1 wild-type controls. RESULTS: We report a specific gene expression signature of 735 up-regulated and 473 down-regulated genes in IKZF1 deleted primary B-ALL pediatric patient samples. Gene ontology studies revealed an up-regulation of genes associated with cell adhesion, cytoskeletal regulation, and motility in IKZF deleted patient samples. Validated up-regulated target genes in IKZF1 deleted patient samples included CTNND1 and PVRL2 (P = 0.0003 and P = 0.001), and RAB3IP and SPIB (P = 0.005 and P = 0.032) were down-regulated. In further studies in IKZF1 knockdown cell lines, apoptosis assays showed no significant chemoresistance. CONCLUSION: IKZF1 knockdown alone does not impart intrinsic chemotherapy resistance suggesting that the association with a poor prognosis may be due to additional lesions, microenvironmental interactions with the bone marrow niche, or other factors. Pediatr Blood Cancer (c) 2014 Wiley Periodicals, Inc.
PMCID:4217284
PMID: 24976218
ISSN: 1545-5009
CID: 1065572
Wnt inhibition leads to improved chemosensitivity in paediatric acute lymphoblastic leukaemia
Dandekar, Smita; Romanos-Sirakis, Eleny; Pais, Faye; Bhatla, Teena; Jones, Courtney; Bourgeois, Wallace; Hunger, Stephen P; Raetz, Elizabeth A; Hermiston, Michelle L; Dasgupta, Ramanuj; Morrison, Debra J; Carroll, William L
While childhood acute lymphoblastic leukaemia (ALL) is now highly curable, the dismal prognosis for children who relapse warrants novel therapeutic approaches. Previously, using an integrated genomic analysis of matched diagnosis-relapse paired samples, we identified overactivation of the Wnt pathway as a possible mechanism of recurrence. To validate these findings and document whether Wnt inhibition may sensitize cells to chemotherapy, we analysed the expression of activated beta-catenin (and its downstream target BIRC5) using multiparameter phosphoflow cytometry and tested the efficacy of a recently developed Wnt inhibitor, iCRT14, in ALL cell lines and patient samples. We observed increased activation of beta-catenin at relapse in 6/10 patients. Furthermore, treatment of leukaemic cell lines with iCRT14 led to significant downregulation of Wnt target genes and combination with traditional chemotherapeutic drugs resulted in a synergistic decrease in viability as well as a significant increase in apoptotic cell death. Finally, pre-treatment of purified blasts from patients with relapsed leukaemia with the Wnt inhibitor followed by exposure to prednisolone, restored chemosensitivity in these cells. Our results demonstrate that overactivation of the Wnt pathway may contribute to chemoresistance in relapsed childhood ALL and that Wnt-inhibition may be a promising therapeutic approach.
PMCID:4207443
PMID: 24995804
ISSN: 0007-1048
CID: 1066072
Targetable Kinase-Activating Lesions in Ph-like Acute Lymphoblastic Leukemia
Roberts, Kathryn G; Li, Yongjin; Payne-Turner, Debbie; Harvey, Richard C; Yang, Yung-Li; Pei, Deqing; McCastlain, Kelly; Ding, Li; Lu, Charles; Song, Guangchun; Ma, Jing; Becksfort, Jared; Rusch, Michael; Chen, Shann-Ching; Easton, John; Cheng, Jinjun; Boggs, Kristy; Santiago-Morales, Natalia; Iacobucci, Ilaria; Fulton, Robert S; Wen, Ji; Valentine, Marcus; Cheng, Cheng; Paugh, Steven W; Devidas, Meenakshi; Chen, I-Ming; Reshmi, Shalini; Smith, Amy; Hedlund, Erin; Gupta, Pankaj; Nagahawatte, Panduka; Wu, Gang; Chen, Xiang; Yergeau, Donald; Vadodaria, Bhavin; Mulder, Heather; Winick, Naomi J; Larsen, Eric C; Carroll, William L; Heerema, Nyla A; Carroll, Andrew J; Grayson, Guy; Tasian, Sarah K; Moore, Andrew S; Keller, Frank; Frei-Jones, Melissa; Whitlock, James A; Raetz, Elizabeth A; White, Deborah L; Hughes, Timothy P; Guidry Auvil, Jaime M; Smith, Malcolm A; Marcucci, Guido; Bloomfield, Clara D; Mrozek, Krzysztof; Kohlschmidt, Jessica; Stock, Wendy; Kornblau, Steven M; Konopleva, Marina; Paietta, Elisabeth; Pui, Ching-Hon; Jeha, Sima; Relling, Mary V; Evans, William E; Gerhard, Daniela S; Gastier-Foster, Julie M; Mardis, Elaine; Wilson, Richard K; Loh, Mignon L; Downing, James R; Hunger, Stephen P; Willman, Cheryl L; Zhang, Jinghui; Mullighan, Charles G
Background Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is characterized by a gene-expression profile similar to that of BCR-ABL1-positive ALL, alterations of lymphoid transcription factor genes, and a poor outcome. The frequency and spectrum of genetic alterations in Ph-like ALL and its responsiveness to tyrosine kinase inhibition are undefined, especially in adolescents and adults. Methods We performed genomic profiling of 1725 patients with precursor B-cell ALL and detailed genomic analysis of 154 patients with Ph-like ALL. We examined the functional effects of fusion proteins and the efficacy of tyrosine kinase inhibitors in mouse pre-B cells and xenografts of human Ph-like ALL. Results Ph-like ALL increased in frequency from 10% among children with standard-risk ALL to 27% among young adults with ALL and was associated with a poor outcome. Kinase-activating alterations were identified in 91% of patients with Ph-like ALL; rearrangements involving ABL1, ABL2, CRLF2, CSF1R, EPOR, JAK2, NTRK3, PDGFRB, PTK2B, TSLP, or TYK2 and sequence mutations involving FLT3, IL7R, or SH2B3 were most common. Expression of ABL1, ABL2, CSF1R, JAK2, and PDGFRB fusions resulted in cytokine-independent proliferation and activation of phosphorylated STAT5. Cell lines and human leukemic cells expressing ABL1, ABL2, CSF1R, and PDGFRB fusions were sensitive in vitro to dasatinib, EPOR and JAK2 rearrangements were sensitive to ruxolitinib, and the ETV6-NTRK3 fusion was sensitive to crizotinib. Conclusions Ph-like ALL was found to be characterized by a range of genomic alterations that activate a limited number of signaling pathways, all of which may be amenable to inhibition with approved tyrosine kinase inhibitors. Trials identifying Ph-like ALL are needed to assess whether adding tyrosine kinase inhibitors to current therapy will improve the survival of patients with this type of leukemia. (Funded by the American Lebanese Syrian Associated Charities and others.).
PMCID:4191900
PMID: 25207766
ISSN: 0028-4793
CID: 1181572
HLA-DRB1*07:01 is associated with a higher risk of asparaginase allergies
Fernandez, Christian A; Smith, Colton; Yang, Wenjian; Date, Mihir; Bashford, Donald; Larsen, Eric; Bowman, W Paul; Liu, Chengcheng; Ramsey, Laura B; Chang, Tamara; Turner, Victoria; Loh, Mignon L; Raetz, Elizabeth A; Winick, Naomi J; Hunger, Stephen P; Carroll, William L; Onengut-Gumuscu, Suna; Chen, Wei-Min; Concannon, Patrick; Rich, Stephen S; Scheet, Paul; Jeha, Sima; Pui, Ching-Hon; Evans, William E; Devidas, Meenakshi; Relling, Mary V
Asparaginase is a therapeutic enzyme used to treat leukemia and lymphoma, with immune responses resulting in suboptimal drug exposure and a greater risk of relapse. To elucidate whether there is a genetic component to the mechanism of asparaginase-induced immune responses, we imputed human leukocyte antigen (HLA) alleles in patients of European ancestry enrolled on leukemia trials at St. Jude Children's Research Hospital (n = 541) and the Children's Oncology Group (n= 1,329). We identified a higher incidence of hypersensitivity and anti-asparaginase antibodies in patients with HLA-DRB1*07:01 alleles (P = 7.5 x 10-5, OR = 1.64; P = 1.4 x 10-5, OR = 2.92, respectively). Structural analysis revealed that high-risk amino acids were located within the binding pocket of the HLA protein, possibly affecting the interaction between asparaginase epitopes and the HLA-DRB1 protein. Using a sequence-based consensus approach, we predicted the binding affinity of HLA-DRB1 alleles for asparaginase epitopes, and patients whose HLA genetics predicted high-affinity binding had more allergy (P = 3.3 x 10-4, OR = 1.38). Our results suggest a mechanism of allergy whereby HLA-DRB1 alleles that confer high-affinity binding to asparaginase epitopes lead to a higher frequency of reactions. Studies were registered at ClinicalTrials.gov, identifiers: NCT00137111, NCT00549848, NCT00005603, and NCT00075725.
PMCID:4141516
PMID: 24970932
ISSN: 0006-4971
CID: 1051392
A Phase I Study of EZN-3042, a Novel Survivin Messenger Ribonucleic Acid (mRNA) Antagonist, Administered in Combination With Chemotherapy in Children With Relapsed Acute Lymphoblastic Leukemia (ALL): A Report From the Therapeutic Advances in Childhood Leukemia and Lymphoma (TACL) Consortium
Raetz, Elizabeth A; Morrison, Debra; Romanos-Sirakis, Eleny; Gaynon, Paul; Sposto, Richard; Bhojwani, Deepa; Bostrom, Bruce C; Brown, Patrick; Eckroth, Elena; Cassar, Jeannette; Malvar, Jemily; Buchbinder, Aby; Carroll, William L
To address the therapeutic challenges in childhood relapsed ALL, a phase 1 study combining a survivin mRNA antagonist, EZN-3042, with reinduction chemotherapy was developed for pediatric patients with second or greater bone marrow relapses of B-lymphoblastic leukemia. EZN-3042 was administered as a single agent on days -5 and -2 and then in combination with a 4-drug reinduction platform on days 8, 15, 22, and 29. Toxicity and the biological activity of EZN-3042 were assessed. Six patients were enrolled at dose level 1 (EZN-3042 2.5 mg/kg/dose). Two dose-limiting toxicities were observed: 1 patient developed a grade 3 gamma-glutamyl transferase elevation and another patient developed a grade 3 gastrointestinal bleeding. Downmodulation of survivin mRNA and protein were assessed after single-agent dosing and decreased expression was observed in 2 of 5 patients with sufficient material for analysis. Although some biological activity was observed, the combination of EZN-3042 with intensive reinduction chemotherapy was not tolerated at a dose that led to consistent downregulation of survivin expression. The trial was terminated following the completion of dose level 1, after further clinical development of this agent was halted.
PMCID:4238428
PMID: 24276047
ISSN: 1077-4114
CID: 703662
The Biology of Relapsed Acute Lymphoblastic Leukemia: Opportunities for Therapeutic Interventions
Bhatla, Teena; Jones, Courtney L; Meyer, Julia A; Vitanza, Nicholas A; Raetz, Elizabeth A; Carroll, William L
Although great strides have been made in the improvement of outcome for newly diagnosed pediatric acute lymphoblastic leukemia because of refinements in risk stratification and selective intensification of therapy, the prognosis for relapsed leukemia has lagged behind significantly. Understanding the underlying biological pathways responsible for drug resistance is essential to develop novel approaches for the prevention of recurrence and treatment of relapsed disease. High throughput genomic technologies have the potential to revolutionize cancer care in this era of personalized medicine. Using such advanced technologies, we and others have shown that a diverse assortment of cooperative genetic and epigenetic events drive the resistant phenotype. Herein, we summarize results using a variety of genomic technologies to highlight the power of this methodology in providing insight into the biological mechanisms that impart resistant disease.
PMCID:4264573
PMID: 24942023
ISSN: 1077-4114
CID: 1036812
Loss of TBL1XR1 Disrupts Glucocorticoid Receptor Recruitment to Chromatin and Results in Glucocorticoid Resistance in a B-Lymphoblastic Leukemia Model
Jones, Courtney L; Bhatla, Teena; Blum, Roy; Wang, Jinhua; Paugh, Steven W; Wen, Xin; Bourgeois, Wallace; Bitterman, Danielle S; Raetz, Elizabeth A; Morrison, Debra J; Teachey, David T; Evans, William E; Garabedian, Michael J; Carroll, William L
Although great advances have been made in the treatment of pediatric acute lymphoblastic leukemia, up to one out of five patients will relapse and their prognosis thereafter is dismal. We have previously identified recurrent deletions in TBL1XR1, which encodes for an F-box like protein responsible for regulating the nuclear hormone repressor (NCoR) complex stability. Here we model TBL1XR1 deletions in B-precursor ALL cell lines and show TBL1XR1 knockdown results in reduced glucocorticoid receptor recruitment to glucocorticoid responsive genes, and ultimately decreased glucocorticoid signaling caused by increased levels of NCoR1 and HDAC3. Reduction in glucocorticoid signaling in TBL1XR1 depleted lines resulted in resistance to glucocorticoid agonists, but not to other chemotherapeutic agents. Importantly, we show that treatment with the HDAC inhibitor SAHA restores sensitivity to prednisolone in TBL1XR1 depleted cells. Altogether, our data indicates that loss of TBL1XR1 is a novel driver of glucocorticoid-resistance in ALL and that epigenetic therapy may have future application in restoring drug sensitivity at relapse.
PMCID:4110265
PMID: 24895125
ISSN: 0021-9258
CID: 1030982
Long-term follow-up of imatinib in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia: Children's Oncology Group study AALL0031
Schultz, K R; Carroll, A; Heerema, N A; Bowman, W P; Aledo, A; Slayton, W B; Sather, H; Devidas, M; Zheng, H W; Davies, S M; Gaynon, P S; Trigg, M; Rutledge, R; Jorstad, D; Winick, N; Borowitz, M J; Hunger, S P; Carroll, W L; Camitta, B
We previously reported preliminary findings that post induction imatinib mesylate (340 mg/m(2)/day), in combination with intensive chemotherapy, resulted in outcomes similar to blood and marrow transplant (BMT) for pediatric patients with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). We now report 5-year outcomes of imatinib plus intensive chemotherapy in 91 children (1-21 years) with and without allogeneic BMT (N=91). We explore the impacts of additional chromosomal abnormalities and minimal residual disease (MRD) by flow cytometry on outcomes. The 5-year disease-free survival was similar for Cohort 5 patients, treated with chemotherapy plus imatinib (70%+/-12%, n=28), sibling donor BMT patients (65%+/-11%, n=21) and unrelated donor BMT patients (59+/-15%; P=0.60, n=13). Patients with additional cytogenetic abnormalities had worse outcomes (P=0.05). End induction (pre-imatinib) MRD was not prognostic for Cohort 5 or allogeneic BMT patients, although limited by small numbers. The re-induction rate following relapse was similar to other higher-risk ALL groups. Longer-term follow-up confirms our initial observation of substantially good outcomes for children and adolescents with Ph+ ALL treated with imatinib plus intensive chemotherapy with no advantage for allogeneic BMT.
PMCID:4282929
PMID: 24441288
ISSN: 0887-6924
CID: 1131552