32 results on '"Mignon Loh"'
Search Results
2. P329: HIGH IMMUNOPROTEASOME ACTIVITY AS NOVEL INDICATOR FOR SENSITIVITY TO BORTEZOMIB-CONTAINING CHEMOTHERAPY IN ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS FROM CHILDREN’S ONCOLOGY GROUP TRIAL AALL1231
- Author
-
Margot Roeten, Suzanne van Dijk, Johan van Meerloo, Zinia Kwidama, Gaya Jenkins, David Teachey, Meenakshi Devidas, Mignon Loh, Gertjan Kaspers, Sonja Zweegman, Gerrit Jansen, Terzah Horton, and Jacqueline Cloos
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. Clinical parameter-based prediction of DNA methylation classification generates a prediction model of prognosis in patients with juvenile myelomonocytic leukemia
- Author
-
Takahiro Imaizumi, Julia Meyer, Manabu Wakamatsu, Hironobu Kitazawa, Norihiro Murakami, Yusuke Okuno, Taro Yoshida, Daichi Sajiki, Asahito Hama, Seiji Kojima, Yoshiyuki Takahashi, Mignon Loh, Elliot Stieglitz, and Hideki Muramatsu
- Subjects
Medicine ,Science - Abstract
Abstract Juvenile myelomonocytic leukemia (JMML) is a rare heterogeneous hematological malignancy of early childhood characterized by causative RAS pathway mutations. Classifying patients with JMML using global DNA methylation profiles is useful for risk stratification. We implemented machine learning algorithms (decision tree, support vector machine, and naïve Bayes) to produce a DNA methylation-based classification according to recent international consensus definitions using a well-characterized pooled cohort of patients with JMML (n = 128). DNA methylation was originally categorized into three subgroups: high methylation (HM), intermediate methylation (IM), and low methylation (LM), which is a trichotomized classification. We also dichotomized the subgroups as HM/IM and LM. The decision tree model showed high concordances with 450k-based methylation [82.3% (106/128) for the dichotomized and 83.6% (107/128) for the trichotomized subgroups, respectively]. With an independent cohort (n = 72), we confirmed that these models using both the dichotomized and trichotomized classifications were highly predictive of survival. Our study demonstrates that machine learning algorithms can generate clinical parameter-based models that predict the survival outcomes of patients with JMML and high accuracy. These models enabled us to rapidly and effectively identify candidates for augmented treatment following diagnosis.
- Published
- 2022
- Full Text
- View/download PDF
4. Aurora A kinase as a target for therapy in TCF3-HLF rearranged acute lymphoblastic leukemia
- Author
-
Jessica Leonard, Joelle SJ Wolf, Michelle Degnin, Christopher A. Eide, Dorian LaTocha, Kyle Lenz, Beth Wilmot, Charles G. Mullighan, Mignon Loh, Stephen P Hunger, Brian J Druker, Marc M Loriaux, Jeffrey W Tyner, and Bill H Chang
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2021
- Full Text
- View/download PDF
5. KRAS insertion mutations are oncogenic and exhibit distinct functional properties
- Author
-
Yasmine White, Aditi Bagchi, Jessica Van Ziffle, Anagha Inguva, Gideon Bollag, Chao Zhang, Heidi Carias, David Dickens, Mignon Loh, Kevin Shannon, and Ari J. Firestone
- Subjects
Science - Abstract
Amino acid substitutions in K-Ras that constitutively activate the protein are common in cancer. Here, the authors describe mutations in the K-RasSwitch 2 domain and show that the mutant proteins accumulate in the active conformation, exhibit defective binding to PI3 kinase, and are hypersensitive to MEK inhibitors.
- Published
- 2016
- Full Text
- View/download PDF
6. MEK inhibitors for neurofibromatosis type 1 manifestations: Clinical evidence and consensus
- Author
-
Peter M K de Blank, Andrea M Gross, Srivandana Akshintala, Jaishri O Blakeley, Gideon Bollag, Ashley Cannon, Eva Dombi, Jason Fangusaro, Bruce D Gelb, Darren Hargrave, AeRang Kim, Laura J Klesse, Mignon Loh, Staci Martin, Christopher Moertel, Roger Packer, Jonathan M Payne, Katherine A Rauen, Jonathan J Rios, Nathan Robison, Elizabeth K Schorry, Kevin Shannon, David A Stevenson, Elliot Stieglitz, Nicole J Ullrich, Karin S Walsh, Brian D Weiss, Pamela L Wolters, Kaleb Yohay, Marielle E Yohe, Brigitte C Widemann, and Michael J Fisher
- Subjects
Mitogen-Activated Protein Kinase Kinases ,Neurofibroma, Plexiform ,Cancer Research ,low-grade glioma ,Neurofibroma ,Consensus ,Neurofibromatosis 1 ,Reviews ,neurofibromatosis type 1 ,RASopathy ,plexiform neurofibromas ,Plexiform ,Oncology ,Humans ,Neurology (clinical) ,Child ,MEK inhibitors ,Protein Kinase Inhibitors - Abstract
The wide variety of clinical manifestations of the genetic syndrome neurofibromatosis type 1 (NF1) are driven by overactivation of the RAS pathway. Mitogen-activated protein kinase kinase inhibitors (MEKi) block downstream targets of RAS. The recent regulatory approvals of the MEKi selumetinib for inoperable symptomatic plexiform neurofibromas in children with NF1 have made it the first medical therapy approved for this indication in the United States, the European Union, and elsewhere. Several recently published and ongoing clinical trials have demonstrated that MEKi may have potential benefits for a variety of other NF1 manifestations, and there is broad interest in the field regarding the appropriate clinical use of these agents. In this review, we present the current evidence regarding the use of existing MEKi for a variety of NF1-related manifestations, including tumor (neurofibromas, malignant peripheral nerve sheath tumors, low-grade glioma, and juvenile myelomonocytic leukemia) and non-tumor (bone, pain, and neurocognitive) manifestations. We discuss the potential utility of MEKi in related genetic conditions characterized by overactivation of the RAS pathway (RASopathies). In addition, we review practical treatment considerations for the use of MEKi as well as provide consensus recommendations regarding their clinical use from a panel of experts.
- Published
- 2023
7. Supplementary Figure 2 from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Supplementary Figure 2 shows the exome coverage achieved with whole exome sequencing.
- Published
- 2023
8. Supplementary Legend from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Legend for Supplementary Figures, Table and Files
- Published
- 2023
9. Supplementary Figure 5 from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Supplementary Figure 5:Germline TP53 variants in the validation cohort Sanger sequencing-derived electropherograms displaying the germline variant in Exon 4 are shown.
- Published
- 2023
10. Data from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Purpose: Second malignant neoplasms (SMNs) are severe late complications that occur in pediatric cancer survivors exposed to radiotherapy and other genotoxic treatments. To characterize the mutational landscape of treatment-induced sarcomas and to identify candidate SMN-predisposing variants, we analyzed germline and SMN samples from pediatric cancer survivors.Experimental Design: We performed whole-exome sequencing (WES) and RNA sequencing on radiation-induced sarcomas arising from two pediatric cancer survivors. To assess the frequency of germline TP53 variants in SMNs, Sanger sequencing was performed to analyze germline TP53 in 37 pediatric cancer survivors from the Childhood Cancer Survivor Study (CCSS) without any history of a familial cancer predisposition syndrome but known to have developed SMNs.Results: WES revealed TP53 mutations involving p53′s DNA-binding domain in both index cases, one of which was also present in the germline. The germline and somatic TP53-mutant variants were enriched in the transcriptomes for both sarcomas. Analysis of TP53-coding exons in germline specimens from the CCSS survivor cohort identified a G215C variant encoding an R72P amino acid substitution in 6 patients and a synonymous SNP A639G in 4 others, resulting in 10 of 37 evaluable patients (27%) harboring a germline TP53 variant.Conclusions: Currently, germline TP53 is not routinely assessed in patients with pediatric cancer. These data support the concept that identifying germline TP53 variants at the time a primary cancer is diagnosed may identify patients at high risk for SMN development, who could benefit from modified therapeutic strategies and/or intensive posttreatment monitoring. Clin Cancer Res; 23(7); 1852–61. ©2016 AACR.
- Published
- 2023
11. Supplementary Figure 1 from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Supplementary Figure 1: Radiotherapy-induced sarcomas in pediatric cancer survivors Radiation dosimetry, imaging and histology are shown.
- Published
- 2023
12. Supplementary Files from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Supplementary File 1. SNVs for Patient 1's SMN;Supplementary File 2. SNVs for Patient 2's SMN;Supplementary File 3. List of dinucleotide substitutions;Supplementary File 4. TP53 germline sequencing primers
- Published
- 2023
13. Supplementary Figure 4 from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Supplementary Figure 4 displays Copy number alterations and allele frequencies across all chromosomes for SMNs.
- Published
- 2023
14. Supplementary Table 1 from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Supplementary Table 1. Treatment exposures and TP53 Variant status in the Validation cohort Tumor, TP53 variant status (G215C, A639G), and treatment exposure details are shown for each validation sample (VS).
- Published
- 2023
15. Supplementary Figure 3 from Somatic and Germline TP53 Alterations in Second Malignant Neoplasms from Pediatric Cancer Survivors
- Author
-
Jean L. Nakamura, Smita Bhatia, Kevin M. Shannon, Alice O. Nakamura, Joseph F. Costello, Rosanna Wustrack, Leslie L. Robison, Sue Hammond, Joseph P. Neglia, Robert E. Goldsby, Steven G. DuBois, Mignon Loh, Andrew E. Horvai, Ivan V. Smirnoff, Tali Mazor, Philip R. Davidson, Leah Lee, Katharine Yu, Vincent Lavergne, and Amy L. Sherborne
- Abstract
Supplementary Figure 3: Somatic variants in radiation-induced SMNs Frequencies of specific types of base substitutions in SNVs (synonymous and non-synonymous SNVs) pooled from all samples are shown.
- Published
- 2023
16. Supplementary Table 1 from Chromosome 12p Deletions in TEL-AML1 Childhood Acute Lymphoblastic Leukemia Are Associated with Retrotransposon Elements and Occur Postnatally
- Author
-
Ru-Fang Yeh, Patricia Buffler, Mignon Loh, Carmen Marsit, Martyn T. Smith, Luoping Zhang, Sheng Zhong, Mi Zhou, Roland Green, Rebecca Selzer, Michelle Kang, Jerry Hofmann, and Joseph L. Wiemels
- Abstract
Supplementary Table 1 from Chromosome 12p Deletions in TEL-AML1 Childhood Acute Lymphoblastic Leukemia Are Associated with Retrotransposon Elements and Occur Postnatally
- Published
- 2023
17. Palbociclib in Combination with Chemotherapy in Pediatric and Young Adult Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia and Lymphoma: A Children’s Oncology Group Study (AINV18P1)
- Author
-
Elizabeth Raetz, David T. Teachey, Charles Minard, Xiaowei Liu, Robin Norris, Kristina Z. Denic, Joel Reid, Nikki Evensen, Lia Gore, Elizabeth Fox, Mignon Loh, Brenda Weigel, and William Carroll
- Abstract
Background Cyclin D has been shown to play an essential role in acute lymphoblastic leukemia (ALL) initiation and progression, providing rationale for targeting the CDK4/6-cyclin D complex that regulates cell cycle progression. Procedure The Children’s Oncology Group AINV18P1 phase 1 trial evaluated the CDK4/6 inhibitor, palbociclib, in combination with standard four-drug reinduction chemotherapy in children and young adults with relapsed/refractory B- and T-cell lymphoblastic leukemia (ALL) and lymphoma. Palbociclib (50 mg/m /dose) was administered orally once daily for 21 consecutive days, first as a single agent (days 1-3) and subsequently combined with reinduction chemotherapy. This two-part study was designed to determine the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D) followed by an expansion pharmacokinetic (PK) cohort. Results Twelve heavily pretreated patients enrolled, all of whom were evaluable for toxicity. One dose-limiting hematologic toxicity (DLT) occurred at the starting dose of 50 mg/m /dose orally for 21 days. No additional DLTs were observed in the dose determination or PK expansion cohorts and overall rates of grade 3/4 non-hematologic toxicities were comparable to those observed with the chemotherapy platform alone. Five complete responses were observed, two among four patients with T-ALL and three among seven patients with B-ALL. Pharmacokinetic studies showed similar profiles with both liquid and capsule formulations of palbociclib. Conclusions Palbociclib in combination with reinduction chemotherapy was well tolerated with a RP2D of 50 mg/m /day for 21 days. Complete responses were observed among heavily pretreated patients.
- Published
- 2023
18. Abstract LB077: Analysis of indel and structural variant error profiles in deep next generation sequencing data
- Author
-
Ying Shao, Quang Tran, Pandurang Kolekar, Yanling Liu, Andrea McBride, Tyler Jones, Heather Mulder, Lingyun Ji, Benjamin Huang, Soheil Meshinchi, Jeffery Klco, Jinghui Zhang, William Carroll, Mignon Loh, Patrick Brown, John Easton, and Xiaotu Ma
- Subjects
Cancer Research ,Oncology - Abstract
Background: Accurate detection of low frequency mutations is of critical importance in the study of genetic heterogeneity, such as on the detection of minimal residual diseases for leukemias. Our prior work has resulted in successful error suppression for substitutions (10−4-10−5). However, the error profiles of indels and structural variants (SVs) remain elusive. Results: In this work, we generated ultra-deep sequencing data using our previously established dilution models (COLO829) on known somatic indels (n=23) and SVs (n=17). We discovered that the error rate of indels (10−6) and SVs (1000-fold lower than that of SNVs. This finding was fully recapitulated in our analysis of 347 indels and 1248 SVs discovered from a relapsed B-ALL cohort of 103 patients, although homopolymer indels can have high error rates (>1%). We then performed a comprehensive study of homopolymer indels in 361 cancer driver genes by using whole genome data from 1662 healthy donors from the SJLIFE cohort. Our data indicated that the number of repeating units are highly predictive relative to the error rate of homopolymer indels (R2=0.988, p=4.89 × 10−8). Utilizing these insights, we assayed end-of-induction remission samples from 72 B-cell lymphoblastic leukemia patients that relapsed by selecting ~5 somatic clonal SNV/Indel/SV markers, which confirmed that SVs and indels have >10-fold lower error rates than SNVs. Our next generation sequencing (NGS) approach had 44 positive detections (61%) and outperformed the current standard method of clinical flow cytometry (n=37; 51%) for detecting minimal residual disease. The NGS-based method detected 92% of designed markers for samples with MRD>0.3%, and this detection rate dropped to 27% for MRD between 0.1% and 0.01%, indicating the difficulty in recovering mutant molecules when their frequencies are very low. Conclusions: Overall, we established indel and SV error profiles in deep next generation sequencing data enabling superior tumor detection performance at very low burdens, with lower error rates than what is observed for SNVs. Our work will have a significant impact on the clinical diagnosis and monitoring of human cancers and beyond. Citation Format: Ying Shao, Quang Tran, Pandurang Kolekar, Yanling Liu, Andrea McBride, Tyler Jones, Heather Mulder, Lingyun Ji, Benjamin Huang, Soheil Meshinchi, Jeffery Klco, Jinghui Zhang, William Carroll, Mignon Loh, Patrick Brown, John Easton, Xiaotu Ma. Analysis of indel and structural variant error profiles in deep next generation sequencing data [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr LB077.
- Published
- 2023
19. ALL-049 A Phase 1/2 Study to Evaluate Ponatinib With Chemotherapy in Pediatric Patients With Relapsed/Resistant/Intolerant Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia or With T315I Mutation
- Author
-
Lia Gore, Mignon Loh, Ching-Hon Pui, Yousif Matloub, Michael J. Hanley, Jichang Du, Meliessa Hennessy, Muriel Granier, Andrea Biondi, and Lewis Silverman
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
20. EBV-driven lymphoid neoplasms associated with pediatric ALL maintenance therapy
- Author
-
Sarah Elitzur, Ajay Vora, Birgit Burkhardt, Hiroto Inaba, Andishe Attarbaschi, Andre Baruchel, Gabriele Escherich, Brenda Gibson, Hsi-Che Liu, Mignon Loh, Anthony V. Moorman, Anja Möricke, Rob Pieters, Anne Uyttebroeck, Susan Baird, Jack Bartram, Shlomit Barzilai-Birenboim, Sandeep Batra, Miriam Ben-Harosh, Yves Bertrand, Trudy Buitenkamp, Kenneth Caldwell, Ricardo Drut, Ashley V. Geerlinks, Gil Gilad, John Grainger, Stephanie Haouy, Nicholas Heaney, Mary Huang, Danielle Ingham, Zdenka Krenova, Michaela Kuhlen, Thomas Lehrnbecher, Atsushi Manabe, Felix Niggli, Claudia Paris, Shoshana Revel-Vilk, Pierre Rohrlich, Mohamad G. Sinno, Tomasz Szczepanski, Melanie Tamesberger, Rajasekharan Warrier, Matthias Wolfl, Ronit Nirel, Shai Izraeli, Arndt Borkhardt, and Kjeld Schmiegelow
- Subjects
Immunology ,Cell Biology ,Hematology ,ddc:610 ,Biochemistry - Abstract
The development of a second malignancy after the diagnosis of childhood acute lymphoblastic leukemia (ALL) is a rare event. Certain second malignancies have been linked with specific elements of leukemia therapy, yet the etiology of most second neoplasms remains obscure and their optimal management strategies are unclear. This is a first comprehensive report of non-Hodgkin lymphomas (NHLs) following pediatric ALL therapy, excluding stem-cell transplantation. We analyzed data of patients who developed NHL following ALL diagnosis and were enrolled in 12 collaborative pediatric ALL trials between 1980-2018. Eighty-five patients developed NHL, with mature B-cell lymphoproliferations as the dominant subtype (56 of 85 cases). Forty-six of these 56 cases (82%) occurred during or within 6 months of maintenance therapy. The majority exhibited histopathological characteristics associated with immunodeficiency (65%), predominantly evidence of Epstein-Barr virus–driven lymphoproliferation. We investigated 66 cases of post-ALL immunodeficiency-associated lymphoid neoplasms, 52 from our study and 14 additional cases from a literature search. With a median follow-up of 4.9 years, the 5-year overall survival for the 66 patients with immunodeficiency-associated lymphoid neoplasms was 67.4% (95% confidence interval [CI], 56-81). Five-year cumulative risks of lymphoid neoplasm– and leukemia-related mortality were 20% (95% CI, 10.2-30) and 12.4% (95% CI, 2.7-22), respectively. Concurrent hemophagocytic lymphohistiocytosis was associated with increased mortality (hazard ratio, 7.32; 95% CI, 1.62-32.98; P = .01). A large proportion of post-ALL lymphoid neoplasms are associated with an immunodeficient state, likely precipitated by ALL maintenance therapy. Awareness of this underrecognized entity and pertinent diagnostic tests are crucial for early diagnosis and optimal therapy.
- Published
- 2022
21. Poster: ALL-049 A Phase 1/2 Study to Evaluate Ponatinib With Chemotherapy in Pediatric Patients With Relapsed/Resistant/Intolerant Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia or With T315I Mutation
- Author
-
Lia Gore, Mignon Loh, Ching-Hon Pui, Yousif Matloub, Michael J. Hanley, Jichang Du, Meliessa Hennessy, Muriel Granier, Andrea Biondi, and Lewis Silverman
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
22. Poster: ALL-167: A Phase 1/2 Study to Evaluate the Safety and Efficacy of Ponatinib with Chemotherapy in Pediatric Patients with Relapsed, Resistant, or Intolerant Philadelphia Chromosome-Positive (Ph+) Acute Lymphoblastic Leukemia (ALL) or Have the T315I Mutation
- Author
-
Lia Gore, Mignon Loh, Ching-Hon Pui, Michael J. Hanley, Jichang Du, Yousif Matloub, Meliessa Hennessy, Muriel Granier, Andrea Biondi, and Lewis Silverman
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2021
23. Abstract 2118: Non-coding germline GATA3 variants alter chromatin topology and contribute to pathogenesis of acute lymphoblastic leukemia
- Author
-
Hongbo Yang, Hui Zhang, Yu Luan, Tingting Liu, Kathryn Roberts, Mao-xiang Qian, Bo Zhang, Wenjian Yang, Virginia Perez-Andreu, Jie Xu, Sriranga lyyanki, Da Kuang, Shalini Reshmi, Julie Gastier-Foster, Colton Smith, Ching-Hon Pui, William Evans, Stephen Hunger, Leonidas Platanias, Mary Relling, Charles Mullighan, Mignon Loh, Feng Yue, and Jun Yang
- Subjects
Cancer Research ,Cancer ,Biology ,medicine.disease ,Topology ,humanities ,Germline ,Chromatin ,Oncology ,Transcriptional regulation ,medicine ,Chromatin Loop ,Epigenetics ,Enhancer ,Reprogramming - Abstract
Inherited non-coding genetic variants confer significant disease susceptibility in many cancers. However, the molecular processes of by which germline variants contribute to somatic lesions are poorly understood. We performed targeted sequencing in 5,008 patients and identified a key regulatory germline variant in GATA3 strongly associated with Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL). By creating an isogenic cellular model with CRISPR-Cas9 system, we showed that this variant activated a strong enhancer that significantly upregulated GATA3 transcription, which in turn reshaped the global chromatin accessibility and 3D genome organization. Remarkably, this genotype switch induced a chromatin loop between the CRLF2 oncogene and a distal enhancer, similar to the somatically acquired super-enhancer hijacking event in patients. GATA3 genotype-related alterations in transcriptional control and 3D chromatin organization were further validated in Ph-like ALL patients. Finally, we showed that GATA3 directly regulates CRLF2 and potentiates the oncogenic effects of JAK-STAT signaling in leukemogenesis. Altogether, our results provide evidence for a novel mechanism by which a germline non-coding variant contributes to oncogene activation epigenetic regulation and 3D genome reprogramming. Citation Format: Hongbo Yang, Hui Zhang, Yu Luan, Tingting Liu, Kathryn Roberts, Mao-xiang Qian, Bo Zhang, Wenjian Yang, Virginia Perez-Andreu, Jie Xu, Sriranga lyyanki, Da Kuang, Shalini Reshmi, Julie Gastier-Foster, Colton Smith, Ching-Hon Pui, William Evans, Stephen Hunger, Stephen Hunger, Leonidas Platanias, Mary Relling, Charles Mullighan, Mignon Loh, Feng Yue, Jun Yang. Non-coding germline GATA3 variants alter chromatin topology and contribute to pathogenesis of acute lymphoblastic leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2118.
- Published
- 2021
24. Abstract PR14: Polycomb repressive complex 2 inactivation induces primary chemotherapy resistance in T-ALL by upregulating the TRAP1 mitochondrial chaperone
- Author
-
Aries, Ingrid, primary, Chonghaile, Triona Ni, additional, Karim, Salmaan, additional, Balbach, Sebastian, additional, Burns, Melissa, additional, Pouliot, Gayle, additional, Kristen, Stevenson, additional, Neuberg, Donna, additional, Devidas, Meenakshi, additional, Mignon, Loh, additional, Hunger, Stephen, additional, Winter, Stuart, additional, Teachey, David, additional, Rabin, Karen, additional, Dunsmore, Kimberly, additional, Wood, Brent, additional, Silverman, Lewis, additional, Sallan, Stephen, additional, Vlierberghe, Peter Van, additional, Orkin, Stuart H., additional, Letai, Anthony G., additional, and Gutierrez, Alejandro, additional
- Published
- 2017
- Full Text
- View/download PDF
25. A Pilot Study of Intensified PEG-Asparaginase in High-risk Acute Lymphoblastic Leukemia: Children's Oncology Group Study AALL08P1
- Author
-
Andrew J. Carroll, Naomi J. Winick, Brent L. Wood, Vilmarie Rodriguez, Zo Ann E. Dreyer, William L. Carroll, Michael J. Burke, Nyla A. Heerema, Elizabeth A. Raetz, Stephen P. Hunger, John A. Kairalla, Meenakshi Devidas, Mignon Loh, Michael J. Borowitz, Wanda L. Salzer, and Barbara L. Asselin
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Asparaginase ,medicine.medical_specialty ,Adolescent ,Antineoplastic Agents ,Pilot Projects ,Article ,Polyethylene Glycols ,Sepsis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Dosing ,Child ,Pegaspargase ,business.industry ,Induction chemotherapy ,Infant ,Hematology ,Induction Chemotherapy ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Thrombosis ,Regimen ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Pancreatitis ,Female ,business ,medicine.drug - Abstract
AALL08P1 was designed to determine whether biweekly intensified pegaspargase (I-PEG) was feasible and safe in pediatric patients with newly diagnosed high-risk B-precursor lymphoblastic leukemia when given with Children's Oncology Group hemiaugmented BFM therapy. High-risk average (HR-Avg) patients received standard pegaspargase dosing (6 doses), whereas high-risk high (HR-High) patients received I-PEG biweekly from the start of Consolidation until day 1 of Maintenance. Feasibility and safety were defined in advance as ≥65% of patients tolerating at least 8 doses of I-PEG and 90% requiring ≤49 weeks from day 1 of Consolidation to the initiation of Maintenance. Targeted toxicities included allergic reactions, anaphylaxis, pancreatitis, thrombosis, bleeding, central nervous system events, and sepsis. AALL08P1 enrolled 104 patients; 54 were classified as HR-Avg and 30 as HR-High after completion of induction therapy. Only 53% (16/30) of the HR-High patients received ≥8 total doses of I-PEG and 50% (15/30) took ≤49 weeks from start of Consolidation to the initiation of Maintenance. I-PEG did not significantly increase grade 2 to 5 targeted toxicities. I-PEG was not feasible or safe as defined in AALL08P1. Complete assessment of this regimen was limited due to removal of patients from I-PEG regimen and early closure of the study.
- Published
- 2016
26. Genomic characterization of childhood acute lymphoblastic leukemia
- Author
-
Charles Mullighan and Mignon Loh
- Subjects
Genetics ,Microarray ,Genomics ,Hematology ,Cell cycle ,Biology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Article ,Transcriptome ,Leukemia ,Mutation ,medicine ,Animals ,Humans ,Cell Lineage ,Epigenetics ,Child ,Transcription factor ,Childhood Acute Lymphoblastic Leukemia ,Signal Transduction ,Transcription Factors - Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood malignancy and a leading case of childhood cancer death. The last decade has witnessed a transformation in our understanding of the genetic basis of ALL due to detailed integrative genomic profiling of large cohorts of childhood ALL. Initially using microarray based approaches, and more recently with next-generation sequencing, these studies have enabled more precise sub-classification of ALL, and have shown that each ALL entity is characterized by constellations of structural and sequence mutations that typically perturb key cellular pathways including lymphoid development, cell cycle regulation, tumor suppression, Ras- and tyrosine kinase driven signaling, and epigenetic regulation. Importantly, several of the newly identified genetic alterations have entered the clinic to improve diagnosis and risk stratification, and are being pursued as new targets for therapeutic intervention. Studies of ALL have also led the way in dissecting the subclonal heterogeneity of cancer, and have shown that individual patients commonly harbor multiple related but genetically distinct subclones, and that this genetically determined clonal heterogeneity is an important determinant of relapse. In addition, genome-wide profiling has identified inherited genetic variants that influence ALL risk. Ongoing studies are deploying detailed integrative genetic transcriptomic and epigenetic sequencing to comprehensively define the genomic landscape of ALL. This review describes the recent advances in our understanding of the genetics of ALL, with an emphasis on those alterations of key pathogenic or therapeutic importance.
- Published
- 2013
27. Germline Genetic Variation in ETV6 and Predisposition to Childhood Acute Lymphoblastic Leukemia
- Author
-
Takaya Moriyama, Monika Metzger, Gang Wu, Rina Nishii, Maoxiang Qian, Meenakshi Devidas, Wenjian Yang, Emily Quinn, Julie Gastier-Foster, Elizabeth Raetz, Eric C. Larsen, Paul L. Martin, W. Paul Bowman, Naomi J. Winick, Yoshihiro Komada, Elaine R Mardis, Robert Fulton, Ching-Hon Pui, William E. Evans, Jinghui Zhang, Stephen P Hunger, Mary V. Relling, Kim E Nichols, Mignon Loh, and Jun J Yang
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in children, and the etiology of this aggressive cancer is not fully understood. Common germline polymorphisms in lymphoid development genes and tumor suppressor genes have been associated with ALL susceptibility, although most have modest effects. Only a small fraction of ALL cases are thought to be related to congenital genetic disorders and consequently hereditary predisposition is rarely considered in clinical practice. However, a growing number of rare germline genetic mutations have been discovered in familial ALL (e.g., PAX5, TP53), raising the possibility that the proportion of ALL attributable to inherited predisposition may be higher than currently proposed. In particular, germline ETV6 variations were recently reported in families with hereditary thrombocytopenia and dramatically increased susceptibility to hematologic malignancies (Nat Genet 2015 47: 180 and 535). ETV6 is a transcriptional repressor essential for hematopoiesis and is frequently targeted by somatic genomic aberrations in childhood ALL (e.g., the ETV6-RUNX1 fusion). Therefore, we sought to comprehensively identify ALL predisposition variants in ETV6 and to determine the extent to which these variants contribute to childhood ALL risk in general. We first identified a family with three cases of childhood ALL at St. Jude Children's Research Hospital. Whole exome sequencing of this family (mother and 2 daughters with ALL, the unaffected father and 1 unaffected daughter) identified a single variant in ETV6 (p.R359X) in the 3 cases with ALL and also in the healthy daughter. This nonsense variant is predicted to create a stop codon within the ETS domain of ETV6, resulting in a truncated protein without DNA-binding function. This highly damaging variant is likely to be responsible for the ALL predisposition in this family with a high albeit incomplete penetrance. To comprehensively determine the prevalence of ALL-predisposing alleles in ETV6, we performed targeted sequencing of this gene in 4,405 children with newly-diagnosed ALL enrolled on the Children's Oncology Group (COG) AALL0232, P9904, P9905 and P9906 protocols and St. Jude Total Therapy XIIIA, XIIIB and XV studies. We identified a total of 43 germline variants in the exonic regions of ETV6. Thirty-one of the 43 ETV6 variants were defined as "ALL-related" because they were not found or extremely rare in non-ALL populations (N=60,706). These ALL risk variants included 4 nonsense, 21 missense, 1 splice site, and 5 frameshift variants occurring in 35 children (0.79% of ALL cases studied). Fifteen of the 31 ALL-relatedvariants (48.4%) were clustered in the ETS DNA-binding domain of ETV6. We used the combined annotation dependent depletion algorithm (CADD) to predict deleterious effects of each variant. ALL-related ETV6 variants were significantly more likely to be damaging compared to germline variants observed in the non-ALL population (mean CADD phred-like score of 25.6 vs 15.2, respectively, p We next analyzed the relationship between germline risk variants in ETV6 and clinical features of ALL in a subset of 2,021 cases enrolled on St. Jude and COG frontline ALL trials. These cases were comprehensively evaluted for ALL charateristics and representative of the US childhood ALL population. Children with ALL-related ETV6 variants were significantly older at the time of diagnosis than those without these variants (9.5 years vs 6.4 years; P=0.009). The hyperdiploid leukemia karyotype was strikingly overrepresented in ALL cases harboring germline ETV6 risk variants compared to the wildtype group (64.3% vs 26.8%; P=0.0045). In contrast, the frequency of somatic ETV6 -RUNX1 fusion was much lower in cases with ETV6 germline risk variants, compared to cases with wildtype ETV6 (7.1% vs 22.7%), even though this difference did not reach statistical significance. Of note, there was also a trend towards overrepresentation of females in carriers of ALL-related ETV6 variants (71.4% vs 45.7%; P=0.063). In conclusion, our findings indicate that germline ETV6 variations are important determinants for genetic predisposition to childhood ALL. Disclosures Martin: Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Gentium SpA/Jazz Pharmaceuticals: Research Funding. Evans:Prometheus Labs: Patents & Royalties: Royalties from licensing TPMT genotyping. Hunger:Spectrum Pharmaceuticals: Consultancy; Jazz Pharmaceuticals: Consultancy; Merck: Equity Ownership; Sigma Tau: Consultancy.
- Published
- 2015
28. Genomic profiling of B-progenitor acute lymphoblastic leukemia
- Author
-
Charles Mullighan and Mignon Loh
- Subjects
Clinical Biochemistry ,Fusion Proteins, bcr-abl ,Genomics ,Disease ,Biology ,Article ,Ikaros Transcription Factor ,Mice ,Recurrence ,hemic and lymphatic diseases ,Animals ,Humans ,Receptors, Cytokine ,Child ,Childhood Acute Lymphoblastic Leukemia ,Progenitor ,Regulation of gene expression ,Chromosome Aberrations ,B-Lymphocytes ,Gene Expression Profiling ,PAX5 Transcription Factor ,Janus Kinase 1 ,Janus Kinase 2 ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,CREB-Binding Protein ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Oncology ,Child, Preschool ,Mutation ,Cancer research ,PAX5 - Abstract
Childhood acute lymphoblastic leukemia is comprised of multiple subtypes defined by recurring chromosomal alterations that are important events in leukemogenesis and are widely used in diagnosis and risk stratification, yet fail to fully explain the biology of this disease. In the last 5 years, genome-wide profiling of gene expression, structural DNA alterations and sequence variations has yielded important insights into the nature of submicroscopic genetic alterations that define novel subgroups of acute lymphoblastic leukemia (ALL) and that cooperate with known cytogenetic alterations in leukemogenesis. Importantly, several of these alterations are important determinants of risk of relapse and are potential targets for therapeutic intervention. Here, these advances and future directions in the genomic analysis of ALL are discussed.
- Published
- 2011
29. Quantitative RT-PCR for Expression of a Small Subset of Genes Identifies Novel Prognostic Subgroups in High-Risk Pediatric Precursor B-Cell Acute Lymphoblastic Leukemia (HR-ALL): Clinical Applicability of Gene Expression Microarray Data from Children’s Oncology Group Trials
- Author
-
Carla S. Wilson, William L. Carroll, Naomi Winick, I-Ming Chen, Cheryl L. Willman, Richard Vestal, Eric C. Larsen, Huining Kang, Stephen P. Hunger, Gregory H. Reaman, David R. Czuchlewski, Meenakshi Devidas, Elizabeth Raetz, Bradley Rodgers, Kerem Ar, Ali Bourguet-Vincent, Richard C. Harvey, and Mignon Loh
- Subjects
Oncology ,medicine.medical_specialty ,Microarray ,Receiver operating characteristic ,Microarray analysis techniques ,Immunology ,Cell Biology ,Hematology ,Gold standard (test) ,Biology ,Biochemistry ,Gene expression profiling ,Real-time polymerase chain reaction ,PTPRM ,Internal medicine ,TaqMan ,medicine - Abstract
Accurate risk stratification constitutes the fundamental paradigm of treatment in acute lymphoblastic leukemia (ALL), allowing the intensity of therapy to be tailored to the patient’s risk of relapse. We have recently identified 2 known (E2A-PBX1, MLL) and 6 novel cluster groups with varying prognostic significance within a high risk ALL cohort (COG P9906) using gene expression profiling. Key genes predicted membership in these novel cluster groups, including MUC4, GPR110, IGJ (poorest outcome cluster) and CENTG2 and PTPRM (most favorable outcome cluster). The poor outcome cluster had a 4-year relapse free survival (RFS) of 20.9%, while the favorable outcome group had an RFS of 94.7% -- both significantly different from the RFS of 61% for the overall cohort. In the current study, we perform quantitative RT- PCR for expression of these genes (MUC4, GPR110, IGJ, CENTG2 and PTPRM) in tandem with gene expression microarray analysis on a new cohort (n=85) of high risk ALL patients enrolled in COG 0232 to validate the findings of our prior microarray classification, and identify a manageable subset of discriminatory genes amenable to RT-PCR analysis, thus permitting the detection of these prognostically significant groups in a clinical setting. Evaluation of PBX1 expression by RT-PCR was included for further validation. RT-PCR assays were designed to simulate as closely as possible testing conditions consistent with a clinical assay; thus, RT-PCR was performed using 10 ng of RNA from each sample, run in duplicate in a quantitative TaqMan assay, and normalized to EEF2 expression. The expression of the analyzed genes by RT-PCR showed excellent correlation with the data generated by expression microarray analysis; using receiver operator curve (ROC) analysis and employing the microarray clusters as a gold standard, the analyzed genes produced areas under the curve (AUC) ranging from 0.88 to 1.0 (a finding characteristic of useful clinical tests) and highly desirable sensitivity and specificity for the identification of poor and favorable outcome groups (see table). Cluster analysis using only RT-PCR expression of these six genes reproduced the significant cluster assignments determined by the microarray analysis. RT-PCR expression of the genes showed a wide dynamic range, a valuable feature for a potential clinical assay. Moreover, using a decision tree approach, we established a step-wise algorithm using the RT-PCR data that permits accurate classification of patients into the poor, favorable and neutral outcome groups. Finally, PBX1 was significantly expressed exclusively in patients found to have E2A-PBX1 by cytogenetic or standard PCR analyses. These results confirm our prior microarray findings and demonstrate that the previously-described prognostic subgroups within high-risk pediatric ALL may be identified in a real-time, clinical setting with a robust quantitative RT-PCR assay for expression of as few as two or three genes. Identification of these prognostic subgroups may improve risk classification in ALL, enhance therapeutic targeting, and thereby improve overall outcome. Gene Prognostic group Area under ROC curve p value for ROC curve Sensitivity (%) Specificity (%) MUC4 Poor 0.88
- Published
- 2008
30. Dysregulated RasGRP1 Responds to Cytokine Receptor Input in T Cell Leukemogenesis.
- Author
-
Hartzell, Catherine, Ksionda, Olga, Lemmens, Ed, Coakley, Kristen, Ming Yang, Dail, Monique, Harvey, Richard C., Govern, Christopher, Bakker, Jeroen, Lenstra, Tineke L., Ammon, Kristin, Boeter, Anne, Winter, Stuart S., Mignon Loh, Shannon, Kevin, Chakraborty, Arup K., Wabl, Matthias, and Roose, Jeroen P.
- Published
- 2013
- Full Text
- View/download PDF
31. ARID5B Genetic Polymorphisms Contribute to Racial Disparities in the Incidence and Treatment Outcome of Childhood Acute Lymphoblastic Leukemia.
- Author
-
Heng Xu, Cheng Cheng, Devidas, Meenakshi, Deqing Pei, Yiping Fan, Wenjian Yang, Neale, Geoff, Scheet, Paul, Burchard, Esteban G., Torgerson, Dara G., Eng, Celeste, Dean, Michael, Antillon, Frederico, Winick, Naomi J., Martin, Paul L., Willman, Cheryl L., Camitta, Bruce M., Reaman, Gregory H., Carroll, William L., and Mignon Loh
- Published
- 2012
- Full Text
- View/download PDF
32. Targeting survivin overcomes drug resistance in acute lymphoblastic leukemia.
- Author
-
Park, Eugene, Eun Ji Gang, Yao-Te Hsieh, Schaefer, Paul, Chae, Sanna, Klemm, Lars, Huantes, Sandra, Mignon Loh, Conway, Edward M., Eun-Suk Kang, Hong Hoe Koo, Hofmann, Wolf-Karsten, Heisterkamp, Nora, Pelus, Louis, Keerthivasan, Ganesan, Crispino, John, Kahn, Michael, Müschen, Markus, and Yong-Mi Kim
- Subjects
- *
DRUG resistance in cancer cells , *HEALTH outcome assessment , *LYMPHOBLASTIC leukemia , *GENE targeting , *APOPTOSIS , *GENETICS ,DISEASE relapse prevention - Abstract
Relapse of drug-resistant acute lymphoblastic leukemia (ALL) has been associated with increased expression of survivin/BIRC5, an inhibitor of apoptosis protein, suggesting a survival advantage for ALL cells. In the present study, we report that inhibition of survivin in patient-derived ALL can eradicate leukemia. Targeting survivin with shRNA in combination with chemotherapy resulted in no detectable minimal residual disease in a xenograft model of primary ALL. Similarly, pharmacologic knock-down of survivin using EZN-3042, a novel locked nucleic acid antisense oligonucleotide, in combination with chemotherapy eliminated drug-resistant ALL cells. These findings show the importance of survivin expression in drug resistance and demonstrate that survivin inhibition may represent a powerful approach to overcoming drug resistance and preventing relapse in patients with ALL. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.