1
|
Grazioli P, Orlando A, Giordano N, Noce C, Peruzzi G, Abdollahzadeh B, Screpanti I, Campese AF. Notch-Signaling Deregulation Induces Myeloid-Derived Suppressor Cells in T-Cell Acute Lymphoblastic Leukemia. Front Immunol 2022; 13:809261. [PMID: 35444651 PMCID: PMC9013886 DOI: 10.3389/fimmu.2022.809261] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/09/2022] [Indexed: 12/28/2022] Open
Abstract
Notch receptors deeply influence T-cell development and differentiation, and their dysregulation represents a frequent causative event in "T-cell acute lymphoblastic leukemia" (T-ALL). "Myeloid-derived suppressor cells" (MDSCs) inhibit host immune responses in the tumor environment, favoring cancer progression, as reported in solid and hematologic tumors, with the notable exception of T-ALL. Here, we prove that Notch-signaling deregulation in immature T cells promotes CD11b+Gr-1+ MDSCs in the Notch3-transgenic murine model of T-ALL. Indeed, aberrant T cells from these mice can induce MDSCs in vitro, as well as in immunodeficient hosts. Conversely, anti-Gr1-mediated depletion of MDSCs in T-ALL-bearing mice reduces proliferation and expansion of malignant T cells. Interestingly, the coculture with Notch-dependent T-ALL cell lines, sustains the induction of human CD14+HLA-DRlow/neg MDSCs from healthy-donor PBMCs that are impaired upon exposure to gamma-secretase inhibitors. Notch-independent T-ALL cells do not induce MDSCs, suggesting that Notch-signaling activation is crucial for this process. Finally, in both murine and human models, IL-6 mediates MDSC induction, which is significantly reversed by treatment with neutralizing antibodies. Overall, our results unveil a novel role of Notch-deregulated T cells in modifying the T-ALL environment and represent a strong premise for the clinical assessment of MDSCs in T-ALL patients.
Collapse
Affiliation(s)
- Paola Grazioli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Andrea Orlando
- Department of Molecular Medicine, Sapienza University, Rome, Italy.,Center for Life Nano- and Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Nike Giordano
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Claudia Noce
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nano- and Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | | | | | | |
Collapse
|
2
|
Mengxuan S, Fen Z, Runming J. Novel Treatments for Pediatric Relapsed or Refractory Acute B-Cell Lineage Lymphoblastic Leukemia: Precision Medicine Era. Front Pediatr 2022; 10:923419. [PMID: 35813376 PMCID: PMC9259965 DOI: 10.3389/fped.2022.923419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/02/2022] [Indexed: 12/05/2022] Open
Abstract
With the markedly increased cure rate for children with newly diagnosed pediatric B-cell acute lymphoblastic leukemia (B-ALL), relapse and refractory B-ALL (R/R B-ALL) remain the primary cause of death worldwide due to the limitations of multidrug chemotherapy. As we now have a more profound understanding of R/R ALL, including the mechanism of recurrence and drug resistance, prognostic indicators, genotypic changes and so on, we can use newly emerging technologies to identify operational molecular targets and find sensitive drugs for individualized treatment. In addition, more promising and innovative immunotherapies and molecular targeted drugs that are expected to kill leukemic cells more effectively while maintaining low toxicity to achieve minimal residual disease (MRD) negativity and better bridge hematopoietic stem cell transplantation (HSCT) have also been widely developed. To date, the prognosis of pediatric patients with R/R B-ALL has been enhanced markedly thanks to the development of novel drugs. This article reviews the new advancements of several promising strategies for pediatric R/R B-ALL.
Collapse
Affiliation(s)
- Shang Mengxuan
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Fen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Runming
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Decker CW, Garcia J, Gatchalian K, Arceneaux D, Choi C, Han D, Hernandez JB. Mitofusin-2 mediates doxorubicin sensitivity and acute resistance in Jurkat leukemia cells. Biochem Biophys Rep 2020; 24:100824. [PMID: 33204855 PMCID: PMC7648112 DOI: 10.1016/j.bbrep.2020.100824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022] Open
Abstract
Mitochondria oscillate along a morphological continuum from fragmented individual units to hyperfused tubular networks. Their position at the junction of catabolic and anabolic metabolism couples this morphological plasticity, called mitochondrial dynamics, to larger cellular metabolic programs, which in turn implicate mitochondria in a number of disease states. In many cancers, fragmented mitochondria engage the cell with the biosynthetic capacity of aerobic glycolysis in service of proliferation and progression. Chemo-resistant cancers, however, favor remodeling dynamics that yield fused mitochondrial assemblies utilizing oxidative phosphorylation (OXPHOS) through the electron transport chain (ETC). In this study, expression of Mitofusin-2 (MFN-2), a GTPase protein mediator of mitochondrial fusion, was found to closely correlate to Jurkat leukemia cell survival post doxorubicin (DxR) assault. Moreover, this was accompanied by dramatically increased expression of OXPHOS respiratory complexes and ATP Synthase, as well as a commensurate escalation of state III respiration and respiratory control ratio (RCR). Importantly, CRISPR knockout of MFN-2 resulted in a considerable decrease of doxorubicin (DxR) median lethal dose compared to a treated wildtype control, suggesting an important role of mitochondrial fusion in chemotherapy sensitivity and acute resistance. Doxorubicin induces mitochondrial fusion in surviving jurkat cells Fused mitochondria in surviving cells increase respiration and mitochondria coupling Mitofusin-2 knockout sensitizes cells to doxorubicin
Collapse
Affiliation(s)
- Carl W Decker
- Keck Graduate Institute, Henry E. Riggs School of Applied Life Sciences, 535 Watson Drive, Claremont, CA, 91711, USA
| | - Jerome Garcia
- University of La Verne, Department of Biology, 1950 3rd Street, La Verne, CA, 91750, USA
| | - Kristelle Gatchalian
- Keck Graduate Institute, Henry E. Riggs School of Applied Life Sciences, 535 Watson Drive, Claremont, CA, 91711, USA
| | | | - Clarice Choi
- Keck Graduate Institute, Henry E. Riggs School of Applied Life Sciences, 535 Watson Drive, Claremont, CA, 91711, USA
| | - Derick Han
- Keck Graduate Institute, Department of Pharmaceutical Sciences, School of Pharmacy and Health Sciences, 535 Watson Drive, Claremont, CA, 91711, USA
| | - Jeniffer B Hernandez
- Keck Graduate Institute, Department of Pharmaceutical Sciences, School of Pharmacy and Health Sciences, 535 Watson Drive, Claremont, CA, 91711, USA
| |
Collapse
|
4
|
Qian JJ, Hu X, Wang Y, Zhang Y, Du J, Yang M, Tong H, Qian WB, Wei J, Yu W, Lou YJ, Mao L, Tao Meng H, You LS, Wang L, Li X, Huang X, Cao LH, Zhao JZ, Yan Yan X, Chen YB, Chen Y, Zhang SJ, Jin J, Hu J, Zhu HH. CAG regimen for refractory or relapsed adult T-cell acute lymphoblastic leukemia: A retrospective, multicenter, cohort study. Cancer Med 2020; 9:5327-5334. [PMID: 32492289 PMCID: PMC7402818 DOI: 10.1002/cam4.3079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/30/2020] [Accepted: 02/22/2020] [Indexed: 01/09/2023] Open
Abstract
Adult patients with relapsed or refractory T‐cell acute lymphoblastic leukemia (R/R‐T‐ALL) have extremely poor prognosis, representing an urgent unmet medical need. Finding an optimal salvage regimen to bridge transplantation is a priority. The CAG (cytarabine, aclarubicin, and G‐CSF) regimen was initially used by one group in China, showing unexpectedly promising results in 11 R/R‐T‐ALL patients. Here, we report the multicenter results of 41 patients who received the CAG regimen as salvage therapy. After one cycle of the CAG regimen, complete remission and partial remission were achieved in 33 (80.5%) and two (4.9%) patients, respectively. Failure to respond was observed in six patients (14.6%). Early T‐cell precursor (ETP) (n = 26) and non‐ETP (n = 15) patients had a similar CR rate (80.8% vs 80.0%, P = .95). Among 41 patients, allo‐HSCT was successfully performed in 27 (66%) patients (22 in CR and 5 in non‐CR). With a median follow‐up time of 12 months, the estimated 2‐year overall survival and event‐free survival were 68.8% (95% CI, 47.3%‐83.0%) and 56.5% (95% CI, 37.1%‐71.9%), respectively. The CAG regimen was well‐tolerated, and no early death occurred. Our multicenter results show that the CAG regimen is highly effective and safe, representing a novel choice for adult patients with R/R‐T‐ALL and providing a better bridge to transplantation.
Collapse
Affiliation(s)
- Jie-Jing Qian
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Xiaoxia Hu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Ying Wang
- Department of Hematology, Shanghai Jiaotong University School of Medicine Affiliated Ruijin Hospital North, Shanghai, China
| | - Yi Zhang
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Juan Du
- Department of Hematology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Min Yang
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Hongyan Tong
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Wen-Bin Qian
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Juying Wei
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Wenjun Yu
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Yin-Jun Lou
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Liping Mao
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Hai Tao Meng
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Liang-Shun You
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Libing Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Shanghai, China
| | - Xia Li
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Xin Huang
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Li-Hong Cao
- Department of Hematology, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Jian-Zhi Zhao
- Department of hematology, Shaoxing Central Hospital, Shaoxing, China
| | - Xiao Yan Yan
- Department of Biostatistics, Peking University Clinical Research Institute, Beijing, China
| | - Yu-Bao Chen
- Department of Hematology, Shanghai Jiaotong University School of Medicine Affiliated Ruijin Hospital North, Shanghai, China
| | - Yu Chen
- Department of Hematology, Shanghai Jiaotong University School of Medicine Affiliated Ruijin Hospital North, Shanghai, China
| | - Su-Jiang Zhang
- Department of Hematology, Shanghai Jiaotong University School of Medicine Affiliated Ruijin Hospital North, Shanghai, China
| | - Jie Jin
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Jiong Hu
- Shanghai Institute of Hematology, Department of Hematology, Blood and Marrow Transplantation Center, Collaborative Innovation Center of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Hu Zhu
- Department of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China.,Institute of Hematology, Zhejiang University, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| |
Collapse
|
5
|
Coccaro N, Anelli L, Zagaria A, Specchia G, Albano F. Next-Generation Sequencing in Acute Lymphoblastic Leukemia. Int J Mol Sci 2019; 20:ijms20122929. [PMID: 31208040 PMCID: PMC6627957 DOI: 10.3390/ijms20122929] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/04/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer and accounts for about a quarter of adult acute leukemias, and features different outcomes depending on the age of onset. Improvements in ALL genomic analysis achieved thanks to the implementation of next-generation sequencing (NGS) have led to the recent discovery of several novel molecular entities and to a deeper understanding of the existing ones. The purpose of our review is to report the most recent discoveries obtained by NGS studies for ALL diagnosis, risk stratification, and treatment planning. We also report the first efforts at NGS use for minimal residual disease (MRD) assessment, and early studies on the application of third generation sequencing in cancer research. Lastly, we consider the need for the integration of NGS analyses in clinical practice for genomic patients profiling from the personalized medicine perspective.
Collapse
Affiliation(s)
- Nicoletta Coccaro
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| | - Luisa Anelli
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| | - Antonella Zagaria
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| | - Giorgina Specchia
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| |
Collapse
|
6
|
Abstract
Treatment outcomes for acute lymphoblastic leukemia (ALL), especially pediatric ALL, have greatly improved due to the risk-adapted therapy. Combination of drug development, clinical practice, as well as basic genetic researches has brought the survival rate of ALL from less than 10% to more than 90% today, not only increasing the treatment efficacy but also limiting adverse drug reactions (ADRs). In this review, we summarized the landscape identification of ALL genetic alterations, which provided the opportunity to increase the survival rate and especially minimize the relapse risk of ALL, and highlighted the importance of the development of new technologies of genomic investigation for translational medicine.
Collapse
|
7
|
[Use of blinatumomab in children acute lymphoblastic leukemia in the Grand Ouest interregion: A chance for all]. Bull Cancer 2019; 106:206-215. [PMID: 30638898 DOI: 10.1016/j.bulcan.2018.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/06/2018] [Accepted: 11/14/2018] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Relapsed/refractory acute lymphoblastic leukemia (ALL) in children has a pejorative prognosis and justifies to be treated by hematopoietic stem cell transplantation (HSCT). A minimal residual disease (MRD) before transplantation is a major part of prognosis. Blinatumomab, a bispecific antibody CD19+/CD3+, allowed to achieve a cytologic and molecular complete remission in adults with refractory B-precursor ALL. This retrospective study analyses results from a pediatric cohort treated by blinatumomab thanks to an interregional structuring consortium. PATIENTS AND METHODS Patients between 0 and 23 years old, from the 7 centers of the french "Grand Ouest" interregional network, treated by blinatumomab for a relapsed or refractory ALL, from January 2015 to January 2018, were included. The efficiency of blinatumomab was assessed in terms of complete remission, minimal residual disease, overall survival, and tolerability of treatment. RESULTS Thirteen of 18 patients achieved a complete remission, with negative minimal residual disease for ten of them. Fourteen patients proceeded to stem cell transplantation,. Eight out of 14 patients obtained long term remission after HSCT. As far as tolerance is concerned, no serious adverse event, neurological or psychiatric disorder, was observed. CONCLUSION Thanks to an interregional network collaboration, all children with high risk ALL coming from the western french interregion could be treated by blinatumomab. Blinatumomab offered good hematological conditions to undergo HSCT with a good tolerability.
Collapse
|
8
|
Haploidentical Donor Transplantation Using a Novel Clofarabine-containing Conditioning Regimen for Very High-risk Hematologic Malignant Neoplasms. J Pediatr Hematol Oncol 2018; 40:e479-e485. [PMID: 29750747 PMCID: PMC6197927 DOI: 10.1097/mph.0000000000001222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recurrent/refractory hematologic malignancies have a poor prognosis, and there is a need for novel treatment regimens that can be tolerated by this heavily pretreated patient group. Clofarabine has antileukemic activity with an acceptable toxicity profile. In a phase I clinical trial (NCT00824135), we substituted clofarabine for fludarabine in a well-established reduced-intensity conditioning regimen for a T cell-depleted, mismatched-related (haploidentical) donor transplant backbone and explored the maximum tolerated dose of clofarabine in this combination in 15 patients undergoing hematopoietic cell transplantation for recurrent/refractory or secondary leukemia. Clofarabine was well tolerated at a dose of 50 mg/m/d for 5 days in this regimen, with minimal treatment-related mortality in a heavily pretreated group of high-risk patients. All patients exhibited quick hematopoietic recovery, with median times to neutrophil and platelet engraftment being 11 and 16 days, respectively. Transient elevation of transaminases was the most common toxicity-observed in 13 patients (86.7%), with 6 (40%) grade III or above. Three patients (20%) developed hepatic veno-occlusive disease. Eleven patients (73.3%) died, with the most common cause of death being disease relapse (in 9 patients [60%]), followed by treatment-related mortality (in 2 patients [13.3%]). Four (26.6%) of the patients are long-term survivors.
Collapse
|
9
|
Cheng H, Huang CM, Wang Y, Hu XX, Xu XQ, Song XM, Tang GS, Chen L, Yang JM. Microarray profiling and co-expression network analysis of the lncRNAs and mRNAs associated with acute leukemia in adults. MOLECULAR BIOSYSTEMS 2018; 13:1102-1108. [PMID: 28428987 DOI: 10.1039/c6mb00874g] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) are common types of acute leukemia in adults and cause low survival rate and poor outcome after 5 years despite high rates of complete remission (CR) with modern chemotherapeutic regimens. To understand the distinct mechanisms in leukemogenesis for ALL and AML and to identify markers for diagnosis and treatment, lncRNA and mRNA expression profiles of AML and ALL patients and healthy controls were generated using microarray analysis. For comparison, the differentially expressed mRNA functions were annotated using gene ontology (GO) and pathway analysis. The microarray revealed that 1011 lncRNAs and 2656 mRNAs differed in AML patients and 6069 lncRNAs and 5338 mRNAs differed in ALL patients from those in healthy controls. The GO terms and KEGG pathway annotation data revealed that the olfactory receptor activity, G-protein coupled receptor activity and olfactory transduction-related genes were significantly associated with AML and ALL. Co-expression network analysis indicated that 108 lncRNAs and 85 mRNAs were included in the co-expression network. This study is the first to explore genome-wide lncRNA expression and co-expression with mRNA patterns in AML and ALL using microarray technology and could provide basic information for new biomarkers or treatment targets to alleviate AML and ALL.
Collapse
Affiliation(s)
- Hui Cheng
- Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Yadav BD, Samuels AL, Wells JE, Sutton R, Venn NC, Bendak K, Anderson D, Marshall GM, Cole CH, Beesley AH, Kees UR, Lock RB. Heterogeneity in mechanisms of emergent resistance in pediatric T-cell acute lymphoblastic leukemia. Oncotarget 2018; 7:58728-42. [PMID: 27623214 PMCID: PMC5312271 DOI: 10.18632/oncotarget.11233] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/28/2016] [Indexed: 11/25/2022] Open
Abstract
Relapse in pediatric T-cell acute lymphoblastic leukemia (T-ALL) remains a significant clinical problem and is thought to be associated with clonal selection during treatment. In this study we used an established pre-clinical model of induction therapy to increase our understanding of the effect of engraftment and chemotherapy on clonal selection and acquisition of drug resistance in vivo. Immune-deficient mice were engrafted with patient diagnostic specimens and exposed to a repeated combination therapy consisting of vincristine, dexamethasone, L-asparaginase and daunorubicin. Any re-emergence of disease following therapy was shown to be associated with resistance to dexamethasone, no resistance was observed to the other three drugs. Immunoglobulin/T-cell receptor gene rearrangements closely matched those in respective diagnosis and relapse patient specimens, highlighting that these clonal markers do not fully reflect the biological changes associated with drug resistance. Gene expression profiling revealed the significant underlying heterogeneity of dexamethasone-resistant xenografts. Alterations were observed in a large number of biological pathways, yet no dominant signature was common to all lines. These findings indicate that the biological changes associated with T-ALL relapse and resistance are stochastic and highly individual, and underline the importance of using sophisticated molecular techniques or single cell analyses in developing personalized approaches to therapy.
Collapse
Affiliation(s)
- Babasaheb D Yadav
- Leukaemia Biology Program, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Amy L Samuels
- Division of Children's Leukaemia and Cancer Research, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Julia E Wells
- Division of Children's Leukaemia and Cancer Research, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Rosemary Sutton
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicola C Venn
- Molecular Diagnostics, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Katerina Bendak
- Leukaemia Biology Program, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Denise Anderson
- Division of Bioinformatics and Biostatistics, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Glenn M Marshall
- Kids Cancer Centre, Sydney Children's Hospital, Sydney, New South Wales, Australia
| | - Catherine H Cole
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | - Alex H Beesley
- Division of Children's Leukaemia and Cancer Research, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Ursula R Kees
- Division of Children's Leukaemia and Cancer Research, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Richard B Lock
- Leukaemia Biology Program, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
11
|
Yang F, Fang H, Wang D, Chen Y, Zhai Y, Zhou BBS, Li H. HPRT1 activity loss is associated with resistance to thiopurine in ALL. Oncotarget 2017; 9:2268-2278. [PMID: 29416770 PMCID: PMC5788638 DOI: 10.18632/oncotarget.23405] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/03/2017] [Indexed: 11/26/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is an aggressive hematological tumor resulting from the malignant transformation of lymphoid progenitors. Thiopurine is a widely used drug in the maintaining treatment of ALL. After a period of chemotherapy, 20% of pediatric patients and over 50% of adult patients will relapse. To investigate the mechanisms of drug resistance in vitro, we established the thiopurine resistant cell lines Reh-6MPR (6-MP Resistant cell) and Reh-6TGR (6-TG Resistant cell) by stepwise selection of the ALL cell line Reh. Cell viability assay revealed that 6MPR and 6TGR cells were almost 1000-fold more resistant to thiopurine comparing with the control Reh cells, and thiopurine conversion was significantly impaired in the resistant cells. Mechanistically, a same novel hypoxanthine phosphoribosyl transferase 1 (HPRT1) mutation c.495_496insA (p.V165fs) was found by whole exome sequencing in both resistant cells. The HPRT1 mutation dramaticly decreased the production of [13C5,15N4]-IMP from [13C5,15N4]-hypoxanthine (HX), showed a loss-of-funciton mechanism. Notably, re-expression the wildtype HPRT1 in Reh-6MPR cell can reverse the drug resistance and thiopurine conversion in Reh-6MPR cells. These results highlight the importance of HPRT1's activity in thiopurine resistance.
Collapse
Affiliation(s)
- Fan Yang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China.,Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pediatric Translational Medicine Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Houshun Fang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pediatric Translational Medicine Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Wang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pediatric Translational Medicine Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Chen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pediatric Translational Medicine Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Bin-Bing S Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pediatric Translational Medicine Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Pharmacology and Chemical Biology, School of Basic Medicine and Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Pediatric Translational Medicine Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Pharmacology and Chemical Biology, School of Basic Medicine and Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Feasibility Study of a Novel Experimental Induction Protocol Combining B43-PAP (Anti-CD19) Immunotoxin With Standard Induction Chemotherapy in Children and Adolescents With Relapsed B-Lineage ALL: A Report From the Children's Oncology Group. J Immunother 2016; 38:299-305. [PMID: 26261894 DOI: 10.1097/cji.0000000000000088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND B43-pokeweed antiviral protein (B43-PAP) is a high-affinity anti-CD19 immunotoxin that is capable of causing apoptotic death in B-lineage leukemic cells with a drug-resistant phenotype. B43-PAP exhibited in vivo antileukemic activity in preclinical studies as well as on a single-agent phase I clinical trial. This pediatric phase I/II study evaluated the toxicity profile and efficacy of B43-PAP immunotoxin in combination with standard induction chemotherapy in children and adolescents with relapsed CD19-positive B-lineage acute lymphoblastic leukemia (B-ALL). Pharmacokinetic profile and immunogenicity of B43-PAP were assessed. EXPERIMENTAL DESIGN B43-PAP in combination with standard 3 and 4-drug induction chemotherapy was administered on days 9-13 and 21-25 of a 28-day treatment course with vincristine, prednisone, L-asparaginase, daunomycin, and intrathecal methotrexate. Thirty patients with relapsed B-ALL were enrolled on study CCG-0957. RESULTS Grade III/IV nonhematologic dose-limiting toxicities were encountered in 4 patients evaluable for toxicity and included myalgias, motor dysfunction, pulmonary toxicity, and elevated liver transaminase. Dose-limiting toxicities occurred only with the 4-drug regimen. Fourteen patients achieved a complete remission at the end of induction among the 20 patients evaluable for response. CONCLUSIONS B43-PAP in combination with standard induction chemotherapy can be safely administered and exhibits clinical antileukemic activity against relapsed B-ALL.
Collapse
|
13
|
Wang W, Zimmerman G, Huang X, Yu S, Myers J, Wang Y, Moreton S, Nthale J, Awadallah A, Beck R, Xin W, Wald D, Huang AY, Zhou L. Aberrant Notch Signaling in the Bone Marrow Microenvironment of Acute Lymphoid Leukemia Suppresses Osteoblast-Mediated Support of Hematopoietic Niche Function. Cancer Res 2016; 76:1641-52. [PMID: 26801976 PMCID: PMC4794354 DOI: 10.1158/0008-5472.can-15-2092] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 12/28/2015] [Indexed: 01/01/2023]
Abstract
More than half of T-cell acute lymphoblastic leukemia (T-ALL) patients harbor gain-of-function mutations in the intracellular domain of Notch1. Diffuse infiltration of the bone marrow commonly occurs in T-ALL and relapsed B-cell acute lymphoblastic leukemia patients, and is associated with worse prognosis. However, the mechanism of leukemia outgrowth in the marrow and the resulting biologic impact on hematopoiesis are poorly understood. Here, we investigated targetable cellular and molecular abnormalities in leukemia marrow stroma responsible for the suppression of normal hematopoiesis using a T-ALL mouse model and human T-ALL xenografts. We found that actively proliferating leukemia cells inhibited normal hematopoietic stem and progenitor cell (HSPC) proliferation and homing to the perivascular region. In addition, leukemia development was accompanied by the suppression of the endosteum-lining osteoblast population. We further demonstrated that aberrant Notch activation in the stroma plays an important role in negatively regulating the expression of CXLC12 on osteoblasts and their differentiation. Notch blockade reversed attenuated HSPC cycling, leukemia-associated abnormal blood lineage distribution, and thrombocytopenia as well as recovered osteoblast and HSPC abundance and improved the hematopoietic-supportive functions of osteoblasts. Finally, we confirmed that reduced osteoblast frequency and enhanced Notch signaling were also features of the marrow stroma of human ALL tissues. Collectively, our findings suggest that therapeutically targeting the leukemia-infiltrated hematopoietic niche may restore HSPC homeostasis and improve the outcome of ALL patients.
Collapse
Affiliation(s)
- Weihuan Wang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Grant Zimmerman
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Xiaoran Huang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Shuiliang Yu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Jay Myers
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Yiwei Wang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Stephen Moreton
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Joseph Nthale
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Amad Awadallah
- Department of Pathology, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Rose Beck
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio. Department of Pathology, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Wei Xin
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio. Department of Pathology, University Hospitals Case Medical Center, Cleveland, Ohio
| | - David Wald
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio. Department of Pathology, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Alex Y Huang
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio. Department of Pathology, University Hospitals Case Medical Center, Cleveland, Ohio.
| |
Collapse
|
14
|
Haro S, Tavenard A, Rialland F, Taque S, Guillerm G, Blouin P, Esvan M, Pellier I, Gandemer V. Keep in Mind Quality of Life: Outcome of a Ten-Year Series of Post-Transplantation Early Relapses in Childhood Acute Lymphoblastic Leukemia-A Report from the Grand Ouest Oncology Study Group for Children in France. Biol Blood Marrow Transplant 2016; 22:889-94. [PMID: 26845034 DOI: 10.1016/j.bbmt.2016.01.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/26/2016] [Indexed: 10/22/2022]
Abstract
Relapses of acute lymphoblastic leukemia (ALL) early after hematopoietic stem cell transplantations in children are uncommon but associated with a very poor prognosis. Whereas there are no current recommendations for the management of these relapses, the children's quality of life is an important issue. We studied the outcomes, including 1-year overall survival, complete remission, and quality of life, of 19 children with ALL who relapsed within the first year after their transplantation treated in the 5 participating centers between 2000 and 2011 Patients were distributed as follows: supportive care only (group A), outpatient treatment (mainly steroid and vincristine, group B), or intensive inpatient treatment (group C). There were no significant differences in 1-year overall survival (31.5% for the entire cohort) or remission rate for time between transplantation and relapse (< 6 months or 6 to 12 months), transplantation or disease characteristics, or treatment group. However, time spent in hospital (for treatment and complications) significantly differed between treatment groups B and C (20.8% ± 13.0 versus 59.1% ± 32.9, respectively; P < .05). No differences in organ toxicities, school attendance, or Lansky scores were found between treatment groups. Our sample size-limited data indicate, in a prepersonalized medicine era, that children treated with steroid and vincristine have the same prognosis as those treated with intensive therapy, but they may benefit from improved quality of life. Nevertheless, new therapeutic strategies are required and future prospective trials would help to establish recommendations.
Collapse
Affiliation(s)
- Sophie Haro
- Department of Pediatrics and Genetics, University Hospital of Brest, Brest, France.
| | - Aude Tavenard
- Grand Ouest Oncology Study Group for Children Biostatistic Unit, University Hospital of Rennes, France
| | - Fanny Rialland
- Department of Pediatric Hematology/Oncology, University Hospital of Nantes, Nantes, France
| | - Sophie Taque
- Department of Pediatric Hematology/Oncology, University Hospital of Rennes, Rennes 1 University, France
| | - Gaelle Guillerm
- Department of Clinical Haematology, Institute of Cancerology and Hematology, University Hospital Brest, Brest, France
| | - Pascale Blouin
- Department of Pediatric Onco-Hematology, University Hospital of Tours, Tours, France
| | - Maxime Esvan
- Grand Ouest Oncology Study Group for Children Biostatistic Unit, University Hospital of Rennes, France
| | - Isabelle Pellier
- Department of Pediatric Onco-Hematology, University Hospital of Angers, Angers, France
| | - Virginie Gandemer
- Department of Pediatric Hematology/Oncology, University Hospital of Rennes, Rennes 1 University, France
| |
Collapse
|
15
|
Dai H, Zhang W, Li X, Han Q, Guo Y, Zhang Y, Wang Y, Wang C, Shi F, Zhang Y, Chen M, Feng K, Wang Q, Zhu H, Fu X, Li S, Han W. Tolerance and efficacy of autologous or donor-derived T cells expressing CD19 chimeric antigen receptors in adult B-ALL with extramedullary leukemia. Oncoimmunology 2015; 4:e1027469. [PMID: 26451310 PMCID: PMC4590028 DOI: 10.1080/2162402x.2015.1027469] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/01/2015] [Accepted: 03/03/2015] [Indexed: 01/16/2023] Open
Abstract
The engineering of T lymphocytes to express chimeric antigen receptors (CARs) aims to establish T cell-mediated tumor immunity rapidly. In this study, we conducted a pilot clinical trial of autologous or donor- derived T cells genetically modified to express a CAR targeting the B-cell antigen CD19 harboring 4-1BB and the CD3ζ moiety. All enrolled patients had relapsed or chemotherapy-refractory B-cell lineage acute lymphocytic leukemia (B-ALL). Of the nine patients, six had definite extramedullary involvement, and the rate of overall survival at 18 weeks was 56%. One of the two patients who received conditioning chemotherapy achieved a three-month durable complete response with partial regression of extramedullary lesions. Four of seven patients who did not receive conditioning chemotherapy achieved dramatic regression or a mixed response in the haematopoietic system and extramedullary tissues for two to nine months. Grade 2-3 graft-versus-host disease (GVHD) was observed in two patients who received substantial donor-derived anti-CD19 CART (chimeric antigen receptor-modified T) cells 3-4 weeks after cell infusions. These results show for the first time that donor-derived anti-CD19 CART cells can cause GVHD and regression of extramedullary B-ALL. This study is registered at www.clinicaltrials.gov as NCT01864889.
Collapse
Affiliation(s)
- Hanren Dai
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Wenying Zhang
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Xiaolei Li
- Department of Molecular Biology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Qingwang Han
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Yelei Guo
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Yajing Zhang
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Yao Wang
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| | - Chunmeng Wang
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Fengxia Shi
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Yan Zhang
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Meixia Chen
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Kaichao Feng
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Quanshun Wang
- Department of Hematology; Chinese PLA General Hospital; Beijing, China
| | - Hongli Zhu
- Department of Hematology; Chinese PLA General Hospital; Beijing, China
| | - Xiaobing Fu
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
| | - Suxia Li
- Department of Hematology; Chinese PLA General Hospital; Beijing, China
| | - Weidong Han
- Department of Immunology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
- Department of Bio-therapeutic; Chinese PLA General Hospital; Beijing, China
- Department of Molecular Biology; Institute of Basic Medicine; School of Life Sciences; Chinese PLA General Hospital; Beijing, China
| |
Collapse
|
16
|
Winter SS, Dunsmore KP, Devidas M, Eisenberg N, Asselin BL, Wood BL, Leonard MS, Murphy J, Gastier-Foster JM, Carroll AJ, Heerema NA, Loh ML, Raetz EA, Winick NJ, Carroll WL, Hunger SP. Safe integration of nelarabine into intensive chemotherapy in newly diagnosed T-cell acute lymphoblastic leukemia: Children's Oncology Group Study AALL0434. Pediatr Blood Cancer 2015; 62:1176-83. [PMID: 25755211 PMCID: PMC4433576 DOI: 10.1002/pbc.25470] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/12/2015] [Indexed: 01/26/2023]
Abstract
BACKGROUND Nelarabine has shown impressive single agent clinical activity in T-cell acute lymphoblastic leukemia (T-ALL), but has been associated with significant neurotoxicities in heavily pre-treated patients. We showed previously that it was safe to add nelarabine to a BFM-86 chemotherapy backbone (AALL00P2). Children's Oncology Group (COG) AALL0434 is a Phase III study designed to test the safety and efficacy of nelarabine when incorporated into a COG augmented BFM-based regimen, which increases exposure to agents with potential neurotoxicity compared to the historical AALL00P2 regimen. PROCEDURE AALL0434 included a safety phase to assess nelarabine toxicity. Patients with high-risk (HR) T-ALL were randomized to receive Capizzi-style escalating methotrexate (MTX) plus pegaspargase or high dose (HD) MTX with/without six five-days courses of nelarabine. We report results from 94 patients who participated in the initial safety phase of the study. RESULTS There were no differences in the incidence of peripheral motor neuropathies, sensory neuropathies or central neurotoxicities among those randomized to the nelarabine (n = 47) and non-nelarabine arms (n = 47). CONCLUSIONS The addition of nelarabine to COG-augmented BFM chemotherapy regimen is safe and feasible. The ongoing AALL0434 Efficacy Phase will determine whether the addition of nelarabine treatment improves outcome for patients with T-ALL.
Collapse
Affiliation(s)
- Stuart S. Winter
- University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | | | | | - Nancy Eisenberg
- University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | | | | | | | - John Murphy
- Princess Margaret Hospital for Children, Perth, Australia
| | - Julie M. Gastier-Foster
- Nationwide Children’s Hospital, Columbus, OH 43205,Ohio State University, Columbus, OH 43210
| | | | | | - Mignon L. Loh
- UCSF Medical Center – Parnassus, San Francisco, CA 94143-0106
| | - Elizabeth A. Raetz
- University of Utah, Primary Children’s Hospital, Salt Lake City, UT 84113
| | - Naomi J. Winick
- University of Texas Southwestern Medical Center, Dallas TX 75390-9063
| | - William L. Carroll
- Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016
| | - Stephen P. Hunger
- University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO 80045
| |
Collapse
|
17
|
Myers DE, Yiv S, Qazi S, Ma H, Cely I, Shahidzadeh A, Arellano M, Finestone E, Gaynon PS, Termuhlen A, Cheng J, Uckun FM. CD19-antigen specific nanoscale liposomal formulation of a SYK P-site inhibitor causes apoptotic destruction of human B-precursor leukemia cells. Integr Biol (Camb) 2015; 6:766-80. [PMID: 24910947 DOI: 10.1039/c4ib00095a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We report the anti-leukemic potency of a unique biotargeted nanoscale liposomal nanoparticle (LNP) formulation of the spleen tyrosine kinase (SYK) P-site inhibitor C61. C61-loaded LNP were decorated with a murine CD19-specific monoclonal antibody directed against radiation-resistant CD19-receptor positive aggressive B-precursor acute lymphoblastic leukemia (ALL) cells. The biotargeted C61-LNP were more potent than untargeted C61-LNP and consistently caused apoptosis in B-precursor ALL cells. The CD19-directed C61-LNP also destroyed B-precursor ALL xenograft cells and their leukemia-initiating in vivo clonogenic fraction. This unique nanostructural therapeutic modality targeting the SYK-dependent anti-apoptotic blast cell survival machinery shows promise for overcoming the clinical radiochemotherapy resistance of B-precursor ALL cells.
Collapse
Affiliation(s)
- Dorothea E Myers
- Developmental Therapeutics Program, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles (CHLA), Los Angeles, CA 90027, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW In this article, new insights into the clinical and biological features of paediatric T-lineage acute lymphoblastic leukaemia (ALL) and their impact on treatment outcome have been described. RECENT FINDINGS T-lineage ALL has considerable phenotypic and biological heterogeneity. Compared with B-lineage ALL, the prognostic significance of the presenting white cell count is weaker and the rate of decline in minimal residual disease is slower in patients with T-lineage ALL. Contemporary, response stratified, treatment protocols incorporating dexamethasone have been associated with significant improvements in outcomes and demonstrated that cranial radiotherapy is not essential for preventing central nervous system relapse. Relapse risk remains higher than for B-lineage ALL and outcome after relapse is poor. Early T-precursor phenotype and genetic abnormalities such as activating ABL1 fusions, NOTCH1/FBXW7, and cytosolic 5'-nucleotidase II gene mutations identify patient groups who may benefit from alternative treatment. New agents such as nelarabine, bortezomib, and clofarabine may be effective in preventing unsalvageable relapses identified by slow response to first-line therapy. SUMMARY Around 85% of children and young people with T-lineage ALL are cured by current therapy. Further improvements in outcome can be expected from genetic profile and response-targeted therapeutics.
Collapse
|
19
|
Shalabi H, Angiolillo A, Fry TJ. Beyond CD19: Opportunities for Future Development of Targeted Immunotherapy in Pediatric Relapsed-Refractory Acute Leukemia. Front Pediatr 2015; 3:80. [PMID: 26484338 PMCID: PMC4589648 DOI: 10.3389/fped.2015.00080] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 09/18/2015] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has been used as a targeted approach in cancer therapy. Relapsed and refractory acute leukemia in pediatrics has been difficult to treat with conventional therapy due to dose-limiting toxicities. With the recent success of CD 19 CAR in pediatric patients with B cell acute lymphoblastic leukemia (ALL), this mode of therapy has become a very attractive option for these patients with high-risk disease. In this review, we will discuss current treatment paradigms of pediatric acute leukemia and potential therapeutic targets for additional high-risk populations, including T cell ALL, AML, and infant ALL.
Collapse
Affiliation(s)
- Haneen Shalabi
- Center for Cancer and Blood Disorders, Children's National Medical Center , Washington, DC , USA
| | - Anne Angiolillo
- Center for Cancer and Blood Disorders, Children's National Medical Center , Washington, DC , USA
| | - Terry J Fry
- Hematologic Malignancies Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
20
|
Ceppi F, Cazzaniga G, Colombini A, Biondi A, Conter V. Risk factors for relapse in childhood acute lymphoblastic leukemia: prediction and prevention. Expert Rev Hematol 2014; 8:57-70. [PMID: 25367188 DOI: 10.1586/17474086.2015.978281] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
With current treatment regimens, survival rates for acute lymphoblastic leukemia (ALL) have improved dramatically since the 1980s, with current 5-year overall survival rates estimated at greater than 85%. This success was achieved, in part, through the implementation of risk-stratified therapy. Nevertheless, for a subgroup of patients (15-20%) with newly diagnosed ALL who will ultimately relapse, traditional risk assessment remains inadequate. The risk of relapse may be estimated on the basis of diagnostic features or early treatment response findings. Further progress in this field may thus come from refinement of predictive factors for relapse and treatment adaptation and from the identification of biological subsets of ALL patients who could benefit from specific target therapies. This article summarizes the aspects associated with the identification of predictive factors for relapse in childhood ALL and options available for prevention of disease recurrence.
Collapse
Affiliation(s)
- Francesco Ceppi
- Division of Haematology/Oncology, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
21
|
Woo JS, Alberti MO, Tirado CA. Childhood B-acute lymphoblastic leukemia: a genetic update. Exp Hematol Oncol 2014; 3:16. [PMID: 24949228 PMCID: PMC4063430 DOI: 10.1186/2162-3619-3-16] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 06/03/2014] [Indexed: 01/06/2023] Open
Abstract
In the pediatric population, B-acute lymphoblastic leukemia (B-ALL) is the most prevalent childhood hematological malignancy, as well as the leading cause of childhood cancer-related mortality. Advances in cytogenetics utilizing array-based technologies and next-generation sequencing (NGS) techniques have revealed exciting insights into the genetic basis of this disease, with the hopes of developing individualized treatment plans for affected children. In this comprehensive review, we discuss our current understanding of childhood (pediatric) B-ALL and highlight the most recent genetic advances and their therapeutic implications.
Collapse
Affiliation(s)
- Jennifer S Woo
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, 1010 Veteran Ave, 2nd Floor, room 2212 F, Los Angeles, CA 90024, USA
| | - Michael O Alberti
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, 1010 Veteran Ave, 2nd Floor, room 2212 F, Los Angeles, CA 90024, USA
| | - Carlos A Tirado
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, 1010 Veteran Ave, 2nd Floor, room 2212 F, Los Angeles, CA 90024, USA
| |
Collapse
|
22
|
Zhou F, Xu Y, Qiu Y, Wu X, Zhang Z, Jin R. Ik6 expression provides a new strategy for the therapy of acute lymphoblastic leukemia. Oncol Rep 2014; 31:1373-9. [PMID: 24402122 DOI: 10.3892/or.2014.2969] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 12/16/2013] [Indexed: 11/06/2022] Open
Abstract
Our previous study demonstrated that the dominant-negative Ikaros isoform 6 (Ik6) is overexpressed in Chinese children with newly diagnosed B-acute lymphoblastic leukemia (B-ALL) and is strongly associated with a poor outcome. The purpose of the present study was to further explore the function of Ik6 in B-ALL. The association between Ik6 expression as detected by real-time PCR and efficacy of chemotherapy was evaluated. The effect of the alteration in Ik6 on leukemic cell lines was assessed by in vitro gain-of-function and loss-of-function techniques. PCR analysis showed that Ik6 expression was decreased when patients completed induction chemotherapy and reached complete remission. Ik6 expression was significantly increased when patients suffered relapse. Stable transfection of Ik6 into the Nalm-6 cell line revealed that Ik6 enhanced proliferation of Nalm-6 cells through the promotion of G0/G1-to-S-phase transition and enhanced chemoresistance to chemotherapeutics through anti-apoptotic effects. However, Ik6 expression did not affect the invasion of Nalm-6 cells. In contrast, silencing of Ik6 in Sup-B15 cells significantly inhibited proliferation and increased chemosensitivity. The present study suggests that Ik6 may be a biological marker of chemosensitivity and relapse and Ik6 may provide a potential therapeutic strategy for ALL.
Collapse
Affiliation(s)
- Fen Zhou
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yunyun Xu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yining Qiu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhiquan Zhang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
23
|
Parameswaran R, Lim M, Arutyunyan A, Abdel-Azim H, Hurtz C, Lau K, Müschen M, Yu RK, von Itzstein M, Heisterkamp N, Groffen J. O-acetylated N-acetylneuraminic acid as a novel target for therapy in human pre-B acute lymphoblastic leukemia. ACTA ACUST UNITED AC 2013; 210:805-19. [PMID: 23478187 PMCID: PMC3620349 DOI: 10.1084/jem.20121482] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Removal of 9-O-acetyl residues from the cell surface N-acetylneuraminic acid makes ALL cells drug sensitive. The development of resistance to chemotherapy is a major cause of relapse in acute lymphoblastic leukemia (ALL). Though several mechanisms associated with drug resistance have been studied in detail, the role of carbohydrate modification remains unexplored. Here, we investigated the contribution of 9-O-acetylated N-acetylneuraminic acid (Neu5Ac) to survival and drug resistance development in ALL cells. A strong induction of 9-O-acetylated Neu5Ac including 9-O-acetyl GD3 was detected in ALL cells that developed resistance against vincristine or nilotinib, drugs with distinct cytotoxic mechanisms. Removal of 9-O-acetyl residues from Neu5Ac on the cell surface by an O-acetylesterase made ALL cells more vulnerable to such drugs. Moreover, removal of intracellular and cell surface–resident 9-O-acetyl Neu5Ac by lentiviral transduction of the esterase was lethal to ALL cells in vitro even in the presence of stromal protection. Interestingly, expression of the esterase in normal fibroblasts or endothelial cells had no effect on their survival. Transplanted mice induced for expression of the O-acetylesterase in the ALL cells exhibited a reduction of leukemia to minimal cell numbers and significantly increased survival. This demonstrates that Neu5Ac 9-O-acetylation is essential for survival of these cells and suggests that Neu5Ac de-O-acetylation could be used as therapy to eradicate drug-resistant ALL cells.
Collapse
Affiliation(s)
- Reshmi Parameswaran
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, The Saban Research Institute, Children's Hospital Los Angeles, CA 90089, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ntziachristos P, Mullenders J, Trimarchi T, Aifantis I. Mechanisms of epigenetic regulation of leukemia onset and progression. Adv Immunol 2013; 117:1-38. [PMID: 23611284 DOI: 10.1016/b978-0-12-410524-9.00001-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the past decade, it has become clear that both genetics and epigenetics play pivotal roles in cancer onset and progression. The importance of epigenetic regulation in proper maintenance of cellular state is highlighted by the frequent mutation of chromatin modulating factors across cancer subtypes. Identification of these mutations has created an interest in designing drugs that target enzymes involved in DNA methylation and posttranslational modification of histones. In this review, we discuss recurrent genetic alterations to epigenetic modulators in both myeloid and lymphoid leukemias. Furthermore, we review how these perturbations contribute to leukemogenesis and impact disease outcome and treatment efficacy. Finally, we discuss how the recent advances in our understanding of chromatin biology may impact treatment of leukemia.
Collapse
|
25
|
Abstract
Abstract
Acute lymphoblastic leukemia (ALL) is the most common and one of the most treatable cancers in children. Although the majority of children with ALL are now cured, 10%-20% of patients are predicted to relapse and outcomes with salvage therapy have been disappointing, with approximately only one-third of children surviving long-term after disease recurrence. Several prognostic factors have been identified, with timing of recurrence relative to diagnosis and site of relapse emerging as the most important variables. Despite heterogeneity in the elements of salvage therapy that are delivered in trials conducted internationally, outcomes have been remarkably similar and have remained static. Because most intensive salvage regimens have reached the limit of tolerability, current strategies are focusing on identifying new agents tailored to the unique biology of relapsed disease and identifying methods to develop these agents efficiently for clinical use. Recently, high-resolution genomic analyses of matched pairs of diagnostic and relapse bone marrow samples are emerging as a promising tool for identifying pathways that impart chemoresistance.
Collapse
|
26
|
Dunsmore KP, Devidas M, Linda SB, Borowitz MJ, Winick N, Hunger SP, Carroll WL, Camitta BM. Pilot study of nelarabine in combination with intensive chemotherapy in high-risk T-cell acute lymphoblastic leukemia: a report from the Children's Oncology Group. J Clin Oncol 2012; 30:2753-9. [PMID: 22734022 DOI: 10.1200/jco.2011.40.8724] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PURPOSE Children's Oncology Group study AALL00P2 was designed to assess the feasibility and safety of adding nelarabine to a BFM 86-based chemotherapy regimen in children with newly diagnosed T-cell acute lymphoblastic leukemia (T-ALL). PATIENTS AND METHODS In stage one of the study, eight patients with a slow early response (SER) by prednisone poor response (PPR; ≥ 1,000 peripheral blood blasts on day 8 of prednisone prephase) received chemotherapy plus six courses of nelarabine 400 mg/m(2) once per day; four patients with SER by high minimal residual disease (MRD; ≥ 1% at day 36 of induction) received chemotherapy plus five courses of nelarabine; 16 patients with a rapid early response (RER) received chemotherapy without nelarabine. In stage two, all patients received six 5-day courses of nelarabine at 650 mg/m(2) once per day (10 SER patients [one by MRD, nine by PPR]) or 400 mg/m(2) once per day (38 RER patients; 12 SER patients [three by MRD, nine by PPR]). RESULTS The only significant difference in toxicities was decreased neutropenic infections in patients treated with nelarabine (42% with v 81% without nelarabine). Five-year event-free survival (EFS) rates were 73% for 11 stage one SER patients and 67% for 22 stage two SER patients treated with nelarabine versus 69% for 16 stage one RER patients treated without nelarabine and 74% for 38 stage two RER patients treated with nelarabine. Five-year EFS for all patients receiving nelarabine (n = 70) was 73% versus 69% for those treated without nelarabine (n = 16). CONCLUSION Addition of nelarabine to a BFM 86-based chemotherapy regimen was well tolerated and produced encouraging results in pediatric patients with T-ALL, particularly those with a SER, who have historically fared poorly.
Collapse
Affiliation(s)
- Kimberly P Dunsmore
- University of Virginia Health System, Box 800386, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Burke MJ, Lindgen B, Verneris MR. Treatment of relapsed acute lymphoblastic leukemia: approaches used by pediatric oncologists and bone marrow transplant physicians. Pediatr Blood Cancer 2012; 58:840-5. [PMID: 21796765 DOI: 10.1002/pbc.23269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 06/14/2011] [Indexed: 11/09/2022]
Abstract
BACKGROUND Management of relapsed B-precursor acute lymphoblastic leukemia (ALL) is challenging and varied. We hypothesized that treatment approaches differ between pediatric oncologists and bone marrow transplant (BMT) physicians. PROCEDURE A survey addressing management of relapsed ALL was sent to pediatric oncologists (n = 883) and BMT (n = 86) physicians across North America. RESULTS A number of similarities in treatment approaches were identified including: choice of chemotherapy for re-induction/consolidation, preference for unrelated donors (URDs) in very early marrow relapse and the choice to not use URD donors in late marrow relapse. However, differences between the two disciplines were noted. For patients who relapsed 18-36 months from diagnosis, the majority of oncologists (53.7%) would retreat with chemotherapy while a majority BMT physicians (70.3%) recommended URD transplant (P < 0.001). Oncologists were also less likely to use minimal residual disease (MRD) in relapse assessment compared to BMT physicians (52% vs. 67.2%; P = 0.028) and more oncologists believed MRD testing was experimental and/or not proven in relapsed ALL (27.1% vs. 12.3%; P = 0.011). CONCLUSIONS This study highlights management differences in children with ALL between pediatric oncologists and BMT physicians, identifying opportunities for collaborative clinical trials.
Collapse
Affiliation(s)
- Michael J Burke
- Division of Pediatric Hematology/Oncology, University of Minnesota, Minneapolis, MN, USA.
| | | | | |
Collapse
|
28
|
RALLE pilot: response-guided therapy for marrow relapse in acute lymphoblastic leukemia in children. J Pediatr Hematol Oncol 2012; 34:263-70. [PMID: 22246158 DOI: 10.1097/mph.0b013e3182352da9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite improved treatment results of childhood acute lymphoblastic leukemia (ALL), 20% to 30% have a relapse, and then the outcome is very poor. We studied 40 children with ALL marrow relapse piloting an ALL relapse protocol with well-known drugs and drug combinations by using a concept of response-guided design. We also measured response in logarithmic fashion. Our primary end points were achievement of M1 marrow status, minimal residual disease status below 10, and second remission. The remission induction rate was 90% with 10% induction mortality. After the A blocks (dexamethasone, vincristine, idarubicin and pegylated L-asparaginase), 85% had M1 status, 39% had minimal residual disease ≤1×10, and 66% had 2 to 3 log response. After B1 block (cyclo, VP-16) the figures were 92%, 58%, and 83%, respectively. Twenty-five of 40 patients received allogeneic stem cell transplantation. Three-year event-free survival of the whole cohort was 37%, and the relapse rate was 38%. Three-year event-free survival by risk group was 53% for late, 34% for early, and 21% for very early relapses. An ALL marrow relapse nonresponsive to steroids, vincristine, asparaginase, anthracyclines, and alkylating agents is uncommon, and these classic drugs can still be advocated for induction of ALL relapse. The problems lie in creating a consolidation capable of preventing particularly posttransplant relapses.
Collapse
|
29
|
Parameswaran R, Yu M, Lyu MA, Lim M, Rosenblum MG, Groffen J, Heisterkamp N. Treatment of acute lymphoblastic leukemia with an rGel/BLyS fusion toxin. Leukemia 2012; 26:1786-96. [PMID: 22373785 PMCID: PMC3376225 DOI: 10.1038/leu.2012.54] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy affecting children and a major cause of mortality from hematopoietic malignancies in adults. A substantial number of patients become drug resistant during chemotherapy, necessitating the development of alternative modes of treatment. rGel (recombinant Gelonin)/BlyS (B-lymphocyte stimulator) is a toxin-cytokine fusion protein used for selective killing of malignant B-cells expressing receptors for B-cell-activating factor (BAFF/BLyS) by receptor-targeted delivery of the toxin, Gelonin. Here, we demonstrate that rGel/BLyS binds to ALL cells expressing BAFF receptor (BAFF-R) and upon internalization, it induces apoptosis of these cells and causes downregulation of survival genes even in the presence of stromal protection. Using an immunodeficient transplant model for human ALL, we show that rGel/BLyS prolongs survival of both Philadelphia chromosome-positive and negative ALL-bearing mice. Furthermore, we used AMD3100, a CXCR4 antagonist, to mobilize the leukemic cells protected in the bone marrow (BM) microenvironment and the combination with rGel/BLyS resulted in a significant reduction of the tumor load in the BM and complete eradication of ALL cells from the circulation. Thus, a combination treatment with the B-cell-specific fusion toxin rGel/BLyS and the mobilizing agent AMD3100 could be an effective alternative approach to chemotherapy for the treatment of primary and relapsed ALL.
Collapse
Affiliation(s)
- R Parameswaran
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Clinical and laboratory biology of childhood acute lymphoblastic leukemia. J Pediatr 2012; 160:10-8. [PMID: 21920540 DOI: 10.1016/j.jpeds.2011.08.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 06/28/2011] [Accepted: 08/02/2011] [Indexed: 02/02/2023]
|
31
|
Mahajan A. Guidelines for the management of relapsed acute lymphoblastic leukemia in childhood. APOLLO MEDICINE 2011. [DOI: 10.1016/s0976-0016(11)60011-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
32
|
van den Berg H, de Groot-Kruseman HA, Damen-Korbijn CM, de Bont ESJM, Schouten-van Meeteren AYN, Hoogerbrugge PM. Outcome after first relapse in children with acute lymphoblastic leukemia: a report based on the Dutch Childhood Oncology Group (DCOG) relapse all 98 protocol. Pediatr Blood Cancer 2011; 57:210-6. [PMID: 21337680 DOI: 10.1002/pbc.22946] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 11/08/2010] [Indexed: 12/27/2022]
Abstract
BACKGROUND We report on the treatment of children and adolescents with acute lymphoblastic leukemia (ALL) in first relapse. The protocol focused on: (1) Intensive chemotherapy preceding allogeneic stem cell transplantation (SCT) in early bone marrow relapse; (2) Rotational chemotherapy in late relapse, without donor; (3) Postponement of cerebro-spinal irradiation in late isolated CNS relapse; and (4) Treatment in very late bone marrow relapse with chemotherapy only. METHODS From January 1999 until July 2006 all 158 Dutch pediatric patients with ALL in first relapse were recorded. Ninety-nine patients were eligible; 54 patients with early and 45 with late relapse. Eighteen patients had an isolated extra-medullary relapse; 69 patients had bone marrow involvement only. RESULTS Five-years EFS rates for early and late relapses were 12% and 35%, respectively. For early relapses 5 years EFSs were 25% for patients transplanted; 0% for non-transplanted patients. For late relapses 5 years EFS was 64% for patients treated with chemotherapy only, and 16% for transplanted patients. For very late relapses EFS was 58%. CONCLUSIONS Our data suggest the superiority of SCT for early relapse patients. For late relapses a better outcome is achieved with chemotherapy only using the rotational chemotherapy scheme. The most important factor for survival was interval between first CR and occurrence of the first relapse.
Collapse
Affiliation(s)
- H van den Berg
- Dutch Childhood Oncology Group, The Hague, The Netherlands.
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Ikaros is a zinc-finger transcription factor that plays an important role in the differentiation and proliferation of lymphocytes. Dominant-negative Ikaros isoform 6 (Ik6), one of its common subtypes, is overexpressed in leukemia patients and is associated with unfavorable prognosis in childhood B-cell progenitor acute lymphoblastic leukemia (ALL). This study was to identify specific isoforms, especially Ik6, in Chinese pediatric patients with ALL. The mRNA expression of Ikaros was detected in 88 children with previously untreated ALL by nested reverse transcription-polymerase chain reaction (RT-PCR). Sequencing of the PCR products was performed to identify specific isoforms. The expression of fusion genes was determined by using multiplex RT-PCR. The functional isoforms Ik1, Ik2/3, and dominant negative isoforms Ik4, 6, 8, 9, 10 identified by nested RT-PCR were further confirmed by sequence analysis. In the 88 cases, the Ik6 was found to be overexpressed in 8 of 70 cases of B-lineage ALL and in 1 of 18 cases of T-lineage ALL patients. Among Ik6 B-lineage ALL patients, 3 had expression of BCR/ABL fusion gene and 1 had HOX11 expression. Ik6 overexpression was independent of age, white blood cell count at diagnosis, risk group, and expression of the fusion genes currently measured in China except BCR/ABL (P<0.01). And it was strongly associated with elevated levels of minimal residual disease at day 28 (P<0.01). Ik6 can be included as a high-risk factor at diagnosis. In developing countries with limited resources, it can be economically detected by nested RT-PCR.
Collapse
|
34
|
Hunger SP, Raetz EA, Loh ML, Mullighan CG. Improving outcomes for high-risk ALL: translating new discoveries into clinical care. Pediatr Blood Cancer 2011; 56:984-93. [PMID: 21370430 DOI: 10.1002/pbc.22996] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 12/01/2010] [Indexed: 12/22/2022]
Abstract
High-risk (HR) acute lymphoblastic leukemia (ALL) remains one of the greatest challenges in pediatric oncology. Relapsed ALL is a leading cause of death in young people, and further improvements in outcome will required the development of therapeutic approaches directed against rational therapeutic targets, as escalation of the intensity of existing therapies is limited by toxicity. This review summarizes advances in the biology and treatment of HR and relapsed ALL presented at a symposium at the 2010 American Society for Pediatric Hematology and Oncology Annual Meeting. Analysis of large patient cohorts has identified several factors associated with HR of relapse including older age, T-lineage disease, and persisting minimal residual disease (MRD) early in therapy. As the results of salvage therapy remain poor, new treatment approaches are needed. BCR-ABL1-positive (Ph+) ALL has historically had a very poor outcome, but recent studies have demonstrated the impressive improvements in treatment outcome with the use of tyrosine kinase inhibitors (TKIs). High-resolution genomic profiling of genetic alterations and gene expression has revolutionized our understanding of the genetic basis of ALL, and has identified several alterations associated with poor outcome, including mutations of the lymphoid transcription factor gene IKZF1 (IKAROS), activating mutations of Janus kinases, and rearrangement of the lymphoid cytokine receptor gene CRLF2. These data indicated that the genetic basis of HR-ALL is multifactorial, and have also provided a new potential therapeutic option directed at JAK inhibition.
Collapse
Affiliation(s)
- Stephen P Hunger
- Center for Cancer and Blood Disorders, The Children's Hospital and Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | | | | |
Collapse
|
35
|
Mullighan CG, Willman CL. Advances in the Biology of Acute Lymphoblastic Leukemia-From Genomics to the Clinic. J Adolesc Young Adult Oncol 2011; 1:77-86. [PMID: 23610732 DOI: 10.1089/jayao.2011.0012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite impressive advances in cure rates for childhood acute lymphoblastic leukemia (ALL), ALL remains the leading cause of disease-related death in young people and new therapeutic approaches directed against rational therapeutic targets are urgently required to improve treatment outcomes. This is particularly true for ALL in older children, adolescents, and adults, in whom treatment outcomes are markedly inferior to those of young children. A major goal of current leukemia research is to use comprehensive genomic analysis of the leukemic cell genome, transcriptome, and epigenome to identify critical new genomic alterations that drive leukemogenesis and influence responsiveness to therapy. Genomic analyses in childhood ALL have been remarkably informative and have identified a number of new structural genetic alterations that play important roles in the establishment of the leukemic clone and determine risk of relapse. Notably, many high-risk ALL cases harbor loss-of-function and dominant mutations of genes that encode transcriptional regulators of lymphoid development coupled with mutations that result in activation of cytokine receptor and kinase signaling pathways. These advances have resulted in new diagnostic approaches and therapeutic trials in ALL. This review will discuss these advances and outline challenges for future studies, including the potential role of genome-wide sequencing approaches and the need for detailed studies of the genetics of ALL in the adolescent and young adult population.
Collapse
Affiliation(s)
- Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital , Memphis, Tennessee
| | | |
Collapse
|
36
|
How new advances in genetic analysis are influencing the understanding and treatment of childhood acute leukemia. Curr Opin Pediatr 2011; 23:34-40. [PMID: 21169835 DOI: 10.1097/mop.0b013e3283426260] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review describes the recent advances in genomic profiling that have provided critical new insights into the biology of acute leukemia in children. RECENT FINDINGS Acute leukemia genomes commonly harbor submicroscopic gains and deletions of DNA which target key cellular pathways that influence leukemogenesis and the likelihood of treatment failure, particularly in acute lymphoblastic leukemia (ALL). Notably, genetic alterations targeting transcriptional regulators of lymphoid development are a hallmark of B-progenitor ALL, and alteration of specific genes in this pathway, such as IKZF1 (encoding IKAROS), are associated with high-risk ALL. Integrated genomic profiling has identified potential therapeutic targets in ALL, including aberrant cytokine receptor signaling mediated by rearrangements and mutation of CRLF2 and JAK2. Genome-wide association studies are also providing important insights into the role of inherited genetic variation and susceptibility to ALL. In contrast, genomic profiling of acute myeloid leukemia (AML) has thus far yielded fewer insights, but ongoing resequencing of leukemia genomes is uncovering novel mutations in both ALL and AML. SUMMARY Genomic profiling has identified important new genetic lesions that contribute to leukemogenesis. These findings will have important implications for the development of new diagnostic tests and treatment approaches in high-risk leukemia. Future studies will be increasingly reliant on comprehensive genomic sequencing to reveal the spectrum of genetic alterations in this disease, with the ultimate aim of improving the treatment outcome for leukemia patients.
Collapse
|
37
|
Szczepanek J, Styczyński J, Haus O, Tretyn A, Wysocki M. Relapse of acute lymphoblastic leukemia in children in the context of microarray analyses. Arch Immunol Ther Exp (Warsz) 2011; 59:61-8. [PMID: 21246408 DOI: 10.1007/s00005-010-0110-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 08/19/2010] [Indexed: 10/18/2022]
Abstract
Over the last four decades the treatment of patients with newly diagnosed childhood acute lymphoblastic leukemia (ALL) has improved remarkably. However, still about 20% of children with ALL relapse despite risk-adapted polychemotherapy. The prognosis of relapsed ALL is relatively poor, even with modern aggressive chemotherapy. Identification of the biological and genetic mechanisms contributing to recurrence in patients with ALL is critical for the development of effective therapeutic strategies to treat refractory leukemic patients. Allogeneic hematopoietic stem-cell transplantation is the treatment of choice for many children with relapsed ALL. The gene expression profile obtained by microarray technology could provide important determinants of the drug response and clinical outcome in childhood ALL. Incorporation of the data on expression levels of newly identified genes into existing strategies of risk stratification might improve clinical management. Current microarray data show correlation of in vitro drug resistance with significant patterns of gene expression and explain clinical differences between early and late relapse. Genes involved in cell proliferation, self-renewal and differentiation, protein biosynthesis, carbohydrate metabolism, and DNA replication and repair are usually among those highly expressed in relapsed lymphoblasts. Current status and future perspectives of microarray data on gene expression and drug resistance profile in relapsed pediatric ALL are discussed in this review.
Collapse
Affiliation(s)
- Joanna Szczepanek
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University, Curie-Skłodowskiej 9, 85-094, Bydgoszcz, Poland
| | | | | | | | | |
Collapse
|
38
|
Collins-Underwood JR, Mullighan CG. Genetic Alterations Targeting Lymphoid Development in Acute Lymphoblastic Leukemia. Curr Top Dev Biol 2011; 94:171-96. [DOI: 10.1016/b978-0-12-380916-2.00006-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
39
|
Macy ME, DeRyckere D, Gore L. Vandetanib mediates anti-leukemia activity by multiple mechanisms and interacts synergistically with DNA damaging agents. Invest New Drugs 2010; 30:468-79. [PMID: 21046425 DOI: 10.1007/s10637-010-9572-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 10/20/2010] [Indexed: 12/16/2022]
Abstract
Vandetanib is an orally active small molecule tyrosine kinase inhibitor (TKI) with activity against several pathways implicated in malignancy including the vascular endothelial growth factor receptor pathway, the epidermal growth factor receptor pathway, the platelet derived growth factor receptor β pathway, and REarranged during Transfection pathway. To determine if vandetanib-mediated inhibition of receptor tyrosine kinases is a potential therapeutic strategy for pediatric acute leukemia, these studies aimed to characterize the activity of vandetanib against acute leukemia in vitro. Treatment of leukemia cell lines with vandetanib resulted in a dose-dependent decrease in proliferation and survival. Vandetanib's anti-leukemic activity appeared mediated by multiple mechanisms including accumulation in G1 phase at lower concentrations and apoptosis at higher concentrations. Alterations in cell surface markers also occurred with vandetanib treatment, suggesting induction of differentiation. In combination with DNA damaging agents (etoposide and doxorubicin) vandetanib demonstrated synergistic induction of cell death. However in combination with the anti-metabolite methotrexate, vandetanib had an antagonistic effect on cell death. Although several targets of vandetanib are expressed on acute leukemia cell lines, expression of vandetanib targets did not predict vandetanib sensitivity and alone are therefore not likely candidate biomarkers in patients with acute leukemia. Interactions between vandetanib and standard chemotherapy agents in vitro may help guide choice of combination regimens for further evaluation in the clinical setting for patients with relapsed/refractory acute leukemia. Taken together, these preclinical data support clinical evaluation of vandetanib, in combination with cytotoxic chemotherapy, for pediatric leukemia.
Collapse
Affiliation(s)
- Margaret E Macy
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplantation, University of Colorado Denver, 13123 East 16th Avenue B-115, Aurora, CO 80045, USA.
| | | | | |
Collapse
|
40
|
Beck JC, Cao Q, Trotz B, Smith AR, Weigel BJ, Verneris MR, Burke MJ. Allogeneic hematopoietic cell transplantation outcomes for children with B-precursor acute lymphoblastic leukemia and early or late BM relapse. Bone Marrow Transplant 2010; 46:950-5. [PMID: 20856210 DOI: 10.1038/bmt.2010.217] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Large registry studies have shown superior disease-free survival (DFS) with matched sibling donor (MSD) allogeneic hematopoietic cell transplantation (allo-HCT) over chemotherapy alone for patients with B-precursor acute lymphoblastic leukemia (ALL) and a late BM relapse. As most of these patients will not have an MSD, the decision to pursue an unrelated allo-HCT in second remission (CR2) or await a future relapse and perform HCT in third remission (CR3) continues to be debated. Between 1990 and 2006, 41 children with relapsed B-precursor ALL received a myeloablative allo-HCT at the University of Minnesota. Graft sources consisted of matched related donor (n=11), matched unrelated donor (n=9), and unrelated umbilical cord blood (n=21). Before allo-HCT, 15 patients had an early relapse (<36 months from diagnosis) and 26 had an initial late relapse (36 months from diagnosis). In all, 30 patients (73%) were in CR2 and 11 were in CR3 (27%) at time of allo-HCT. Five year OS/DFS were similar for patients with an early or late marrow relapse, but there was inferior DFS among late-relapse patients transplanted in CR3 compared with CR2 (30% vs 75%, P=0.04). These results suggest that allo-HCT should be pursued in children after a first marrow relapse, rather than waiting for subsequent recurrence.
Collapse
Affiliation(s)
- J C Beck
- Division of Pediatric Hematology/Oncology/Bone and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455 USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Suzuki N, Yumura-Yagi K, Yoshida M, Hara J, Nishimura S, Kudoh T, Tawa A, Usami I, Tanizawa A, Hori H, Ito Y, Miyaji R, Oda M, Kato K, Hamamoto K, Osugi Y, Hashii Y, Nakahata T, Horibe K. Outcome of childhood acute lymphoblastic leukemia with induction failure treated by the Japan Association of Childhood Leukemia study (JACLS) ALL F-protocol. Pediatr Blood Cancer 2010; 54:71-8. [PMID: 19813250 DOI: 10.1002/pbc.22217] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Children with acute lymphoblastic leukemia (ALL) who fail to achieve complete remission (CR) after induction therapy (induction failure: IF) have a poor prognosis; however, there have been few prospective studies in patients with IF. PATIENTS AND METHODS Between April 1997 and March 2005, 27 of 1,237 leukemic patients (2.2%) failed to achieve CR after four- or five-drug induction therapy. Twenty-three of these patients entered the F-protocol study, which mainly consisted of acute-myeloid-leukemia-oriented chemotherapy followed by scheduled hematopoietic cell transplantation (HCT). RESULTS Seventeen (73.9%) of the 23 patients responded to re-induction chemotherapy with CR. Of note, 15 (93.8%) of 16 patients with Philadelphia-chromosome-negative (non-Ph(+)) ALL achieved CR; in contrast, only 2 (28.6%) of 7 Ph(+) patients achieved CR. Fourteen (82.4%) of 17 patients remained in CR (CCR) until their scheduled HCT, 12 of the 14 with CCR underwent HCT as scheduled, and 6 patients remain in first CR after a median of 78 months (range, 49-107 months). The 5-year overall survival (OS) rates of 16 patients with non-Ph(+) and 7 patients with Ph(+) were 43.8 +/- 12.4% and 14.3 +/- 13.2%, respectively (P = 0.012). The 5-year OS rate of the 17 patients who obtained CR by re-induction therapy and the 6 who did not were 47.1 +/- 12.1% and 0%, respectively (P < 0.001). CONCLUSION Acute-myeloid-leukemia-oriented chemotherapy followed by scheduled HCT is a promising treatment strategy for non-Ph(+) ALL patients with IF.
Collapse
Affiliation(s)
- Nobuhiro Suzuki
- Department of Pediatrics, Sapporo Medical University Hospital, Sapporo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ko RH, Ji L, Barnette P, Bostrom B, Hutchinson R, Raetz E, Seibel NL, Twist CJ, Eckroth E, Sposto R, Gaynon PS, Loh ML. Outcome of patients treated for relapsed or refractory acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia Consortium study. J Clin Oncol 2009; 28:648-54. [PMID: 19841326 DOI: 10.1200/jco.2009.22.2950] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Despite improvements in treatment, approximately 20% of patients with acute lymphoblastic leukemia (ALL) experience relapse and do poorly. The Therapeutic Advances in Childhood Leukemia (TACL) Consortium was assembled to assess novel drugs for children with resistant leukemia. We hypothesize that novel agents and combinations that fail to improve baseline complete remission rates in comparable populations are unlikely to contribute to better outcomes and should be abandoned. We sought to define response rates and disease-free survival (DFS) rates in patients treated at TACL institutions, which could serve as a comparator for future studies. PATIENTS AND METHODS We performed a retrospective cohort review of patients with relapsed and refractory ALL previously treated at TACL institutions between the years of 1995 and 2004. Data regarding initial and relapsed disease characteristics, disease response, and survival were collected and compared with those of published reports. RESULTS Complete remission (CR) rates (mean +/- SE) were 83% +/- 4% for early first marrow relapse, 93% +/- 3% for late first marrow relapse, 44% +/- 5% for second marrow relapse, and 27% +/- 6% for third marrow relapse. Five-year DFS rates in CR2 and CR3 were 27% +/- 4% and 15% +/- 7% respectively. CONCLUSION We generally confirm a 40% CR rate for second and subsequent relapse, but our remission rate for early first relapse seems better than that reported in the literature (83% v approximately 70%). Our data may allow useful modeling of an expected remission rate for any population of patients who experience relapse.
Collapse
Affiliation(s)
- Richard H Ko
- Therapeutic Advances in Childhood Leukemia Consortium, Institute for Pediatric Clinical Research and Childrens Center for Cancer and Blood Diseases, University of Southern California-Childrens Hospital Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Genome-wide profiling of genetic alterations in acute lymphoblastic leukemia: recent insights and future directions. Leukemia 2009; 23:1209-18. [DOI: 10.1038/leu.2009.18] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Abstract
While cure rates of over 80% are achieved in contemporary pediatric acute lymphoblastic leukemia (ALL) protocols, most adults with ALL succumb to their disease, and little progress has been made in the treatment of refractory and relapsed ALL. Moreover, the burden of therapy is high in a significant number of newly diagnosed patients, and in all those with relapse. Early response to therapy measured by minimal residual disease evaluation has proven the single most important prognostic factor and is increasingly used in risk stratification. However, as the benefit from intensification of frontline therapy becomes limiting, it becomes increasingly challenging to rescue patients who fail on contemporary risk-adapted protocols. New therapeutic strategies are needed, not only in salvage regimens but also in frontline protocols for patients who are at high risk of relapse. Current novel approaches include new formulations of existing chemotherapeutic agents, new antimetabolites and nucleoside analogs, monoclonal antibodies against leukemic-associated antigens, cellular immunotherapy, and molecular therapeutics. Some have already been adopted into standard regimens, while others remain in early stages of development. This review summarizes the current status of these novel therapies as they get integrated into ALL regimens.
Collapse
Affiliation(s)
- Sima Jeha
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
46
|
Mullighan CG, Su X, Zhang J, Radtke I, Phillips LAA, Miller CB, Ma J, Liu W, Cheng C, Schulman BA, Harvey RC, Chen IM, Clifford RJ, Carroll WL, Reaman G, Bowman WP, Devidas M, Gerhard DS, Yang W, Relling MV, Shurtleff SA, Campana D, Borowitz MJ, Pui CH, Smith M, Hunger SP, Willman CL, Downing JR. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med 2009; 360:470-80. [PMID: 19129520 PMCID: PMC2674612 DOI: 10.1056/nejmoa0808253] [Citation(s) in RCA: 1069] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Despite best current therapy, up to 20% of pediatric patients with acute lymphoblastic leukemia (ALL) have a relapse. Recent genomewide analyses have identified a high frequency of DNA copy-number abnormalities in ALL, but the prognostic implications of these abnormalities have not been defined. METHODS We studied a cohort of 221 children with high-risk B-cell-progenitor ALL with the use of single-nucleotide-polymorphism microarrays, transcriptional profiling, and resequencing of samples obtained at diagnosis. Children with known very-high-risk ALL subtypes (i.e., BCR-ABL1-positive ALL, hypodiploid ALL, and ALL in infants) were excluded from this cohort. A copy-number abnormality was identified as a predictor of poor outcome, and it was then tested in an independent validation cohort of 258 patients with B-cell-progenitor ALL. RESULTS More than 50 recurring copy-number abnormalities were identified, most commonly involving genes that encode regulators of B-cell development (in 66.8% of patients in the original cohort); PAX5 was involved in 31.7% and IKZF1 in 28.6% of patients. Using copy-number abnormalities, we identified a predictor of poor outcome that was validated in the independent validation cohort. This predictor was strongly associated with alteration of IKZF1, a gene that encodes the lymphoid transcription factor IKAROS. The gene-expression signature of the group of patients with a poor outcome revealed increased expression of hematopoietic stem-cell genes and reduced expression of B-cell-lineage genes, and it was similar to the signature of BCR-ABL1-positive ALL, another high-risk subtype of ALL with a high frequency of IKZF1 deletion. CONCLUSIONS Genetic alteration of IKZF1 is associated with a very poor outcome in B-cell-progenitor ALL.
Collapse
Affiliation(s)
- Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Mullighan CG, Phillips LA, Su X, Ma J, Miller CB, Shurtleff SA, Downing JR. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science 2008; 322:1377-80. [PMID: 19039135 PMCID: PMC2746051 DOI: 10.1126/science.1164266] [Citation(s) in RCA: 623] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Most children with acute lymphoblastic leukemia (ALL) can be cured, but the prognosis is dismal for the minority of patients who relapse after treatment. To explore the genetic basis of relapse, we performed genome-wide DNA copy number analyses on matched diagnosis and relapse samples from 61 pediatric patients with ALL. The diagnosis and relapse samples typically showed different patterns of genomic copy number abnormalities (CNAs), with the CNAs acquired at relapse preferentially affecting genes implicated in cell cycle regulation and B cell development. Most relapse samples lacked some of the CNAs present at diagnosis, which suggests that the cells responsible for relapse are ancestral to the primary leukemia cells. Backtracking studies revealed that cells corresponding to the relapse clone were often present as minor subpopulations at diagnosis. These data suggest that genomic abnormalities contributing to ALL relapse are selected for during treatment, and they point to new targets for therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Xiaoping Su
- Department of Pathology, St. Jude Children‘s Research Hospital
| | - Jing Ma
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children‘s Research Hospital
| | | | | | | |
Collapse
|
48
|
Yang JJ, Bhojwani D, Yang W, Cai X, Stocco G, Crews K, Wang J, Morrison D, Devidas M, Hunger SP, Willman CL, Raetz EA, Pui CH, Evans WE, Relling MV, Carroll WL. Genome-wide copy number profiling reveals molecular evolution from diagnosis to relapse in childhood acute lymphoblastic leukemia. Blood 2008; 112:4178-4183. [PMID: 18768390 PMCID: PMC2581992 DOI: 10.1182/blood-2008-06-165027] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Accepted: 08/10/2008] [Indexed: 01/15/2023] Open
Abstract
The underlying pathways that lead to relapse in childhood acute lymphoblastic leukemia (ALL) are unknown. To comprehensively characterize the molecular evolution of relapsed childhood B-precursor ALL, we used human 500K single-nucleotide polymorphism arrays to identify somatic copy number alterations (CNAs) in 20 diagnosis/relapse pairs relative to germ line. We identified 758 CNAs, 66.4% of which were less than 1 Mb, and deletions outnumbered amplifications by approximately 2.5:1. Although CNAs persisting from diagnosis to relapse were observed in all 20 cases, 17 patients exhibited differential CNA patterns from diagnosis to relapse. Of the 396 CNAs observed in 20 relapse samples, only 69 (17.4%) were novel (absent in the matched diagnosis samples). EBF1 and IKZF1 deletions were particularly frequent in this relapsed ALL cohort (25.0% and 35.0%, respectively), suggesting their role in disease recurrence. In addition, we noted concordance in global gene expression and DNA copy number changes (P = 2.2 x 10(-16)). Finally, relapse-specific focal deletion of MSH6 and, consequently, reduced gene expression were found in 2 of 20 cases. In an independent cohort of children with ALL, reduced expression of MSH6 was associated with resistance to mercaptopurine and prednisone, thereby providing a plausible mechanism by which this acquired deletion contributes to drug resistance at relapse.
Collapse
Affiliation(s)
- Jun J Yang
- St Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Factors influencing survival after relapse from acute lymphoblastic leukemia: a Children's Oncology Group study. Leukemia 2008; 22:2142-50. [PMID: 18818707 DOI: 10.1038/leu.2008.251] [Citation(s) in RCA: 442] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite great progress in curing childhood acute lymphoblastic leukemia (ALL), survival after relapse remains poor. We analyzed survival after relapse among 9585 pediatric patients enrolled on Children's Oncology Group clinical trials between 1988 and 2002. A total of 1961 patients (20.5%) experienced relapse at any site. The primary end point was survival. Patients were subcategorized by the site of relapse and timing of relapse from initial diagnosis. Time to relapse remains the strongest predictor of survival. Patients experiencing early relapse less than 18 months from initial diagnosis had a particularly poor outcome with a 5-year survival estimate of 21.0+/-1.8%. Standard risk patients who relapsed had improved survival compared with their higher risk counterparts; differences in survival for the two risk groups was most pronounced for patients relapsing after 18 months. Adjusting for both time and relapse site, multivariate analysis showed that age (10+ years) and the presence of central nervous system disease at diagnosis, male gender, and T-cell disease were significant predictors of inferior post-relapse survival. It can be noted that there was no difference in survival rates for relapsed patients in earlier vs later era trials. New therapeutic strategies are urgently needed for children with relapsed ALL and efforts should focus on discovering the biological pathways that mediate drug resistance.
Collapse
|
50
|
Bailey LC, Lange BJ, Rheingold SR, Bunin NJ. Bone-marrow relapse in paediatric acute lymphoblastic leukaemia. Lancet Oncol 2008; 9:873-83. [PMID: 18760243 DOI: 10.1016/s1470-2045(08)70229-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Marrow relapse is the major obstacle to cure for 10-15% of young patients with acute lymphoblastic leukaemia (ALL). Recent investigations into the biology of minimal residual disease indicate that many early relapses derive from residual cells present at first diagnosis, but some late relapses might represent new mutations in leukaemic cells not eliminated by conventional therapy. Treatment of marrow relapse involves higher doses and more intensive schedules of the drugs used for initial therapy with or without haemopoietic stem cell transplantation. In most reports, transplantation is better than continuation chemotherapy in early marrow relapse, but its role in later relapse is less clear. Current therapy cures 10% of patients with early marrow relapses and 50% of those with late relapses, but outcomes have changed little in the past two decades. Understanding the molecular biology of ALL underlies development of improved risk stratification and new therapies. Although better drugs are needed, introduction of new agents into clinical trials in paediatric disease has been difficult. Innovative trial designs and use of valid surrogate endpoints may expedite this process.
Collapse
Affiliation(s)
- L Charles Bailey
- Division of Oncology, Children's Hospital of Philadelphia, and University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | |
Collapse
|