1
|
Sultan I, Alfaar AS, Sultan Y, Salman Z, Qaddoumi I. Trends in childhood cancer: Incidence and survival analysis over 45 years of SEER data. PLoS One 2025; 20:e0314592. [PMID: 39752445 PMCID: PMC11698462 DOI: 10.1371/journal.pone.0314592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/12/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND The SEER Registry contains U.S. cancer statistics. To assess trends in incidence and survival and the impact of demographic factors among pediatric patients with cancer, we assessed nearly 5 decades (1975-2019) of data. METHODS All patients below the age of 20 with histology-confirmed malignancy were studied. Kaplan-Meier survival curves were generated to evaluate survival trends across treatment periods and ICCC classes. JoinPoint analysis was conducted to identify changes in incidence and survival. RESULTS The incidence of childhood cancer increased from 14.23 cases per 100,000 children in 1975-1979 to 18.89 in 2010-2019, with an average annual percent change of 0.73. This rise was more pronounced in several cancers, including leukemias, lymphomas, brain tumors, hepatic tumors, and gonadal germ cell tumors. Age-adjusted cancer mortality decreased from 4.9 to 2.3 per 100,000. Cancer-related mortality was consistently higher in boys than in girls, and in Black children than in White children. Survival significantly improved, with 5- and 10-year survival rates rising from 63.1% to 85.2% and from 58.8% to 82.7%, respectively. Leukemias showed a substantial increase in 5-year survival from 48.2% ± 1.7% to 85.1% ± 0.4% in 2010-2019. Lymphomas also showed significant improvement, with survival increasing from 72.9% ± 1.7% to 94.2% ± 0.3%. Despite these improvements, the survival of CNS tumors, bone tumors, and sarcomas remained suboptimal, with 5-year survival estimates of approximately 60%. Our joinpoint analysis confirmed our findings but revealed an interesting increase in the incidence of lymphomas limited to the years between 2005 and 2014. CONCLUSION This research elucidates advancements in survival among pediatric patients with cancer. The results offer critical perspectives on pediatric oncology, highlighting the imperative for ongoing innovation in therapeutics. Although the increase in incidence may partially stem from enhanced diagnostic capabilities and more comprehensive registration processes, the underlying causes remain unclear.
Collapse
Affiliation(s)
- Iyad Sultan
- Department of Pediatrics and Artificial Intelligence and Data Innovation Office (AIDI), King Hussein Cancer Center, Amman, Jordan
| | - Ahmad S. Alfaar
- Department of Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yaseen Sultan
- Department of Biostatistics, University of Iowa, Iowa City, IA, United States of America
| | - Zeena Salman
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN, United States of America
| | - Ibrahim Qaddoumi
- Departments of Global Pediatric Medicine and Oncology, St. Jude Children’s Research Hospital, Memphis, TN, United States of America
| |
Collapse
|
2
|
Witek MA, Larkey NE, Bartakova A, Hupert ML, Mog S, Cronin JK, Vun J, August KJ, Soper SA. Microfluidic Affinity Selection of B-Lineage Cells from Peripheral Blood for Minimal Residual Disease Monitoring in Pediatric B-Type Acute Lymphoblastic Leukemia Patients. Int J Mol Sci 2024; 25:10619. [PMID: 39408948 PMCID: PMC11477226 DOI: 10.3390/ijms251910619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Assessment of minimal residual disease (MRD) is the most powerful predictor of outcome in B-type acute lymphoblastic leukemia (B-ALL). MRD, defined as the presence of leukemic cells in the blood or bone marrow, is used for the evaluation of therapy efficacy. We report on a microfluidic-based MRD (MF-MRD) assay that allows for frequent evaluation of blood for the presence of circulating leukemia cells (CLCs). The microfluidic chip affinity selects B-lineage cells, including CLCs using anti-CD19 antibodies poised on the wall of the microfluidic chip. Affinity-selected cells are released from the capture surface and can be subjected to immunophenotyping to enumerate the CLCs, perform fluorescence in situ hybridization (FISH), and/or molecular analysis of the CLCs' mRNA/gDNA. During longitudinal testing of 20 patients throughout induction and consolidation therapy, the MF-MRD performed 116 tests, while only 41 were completed with multiparameter flow cytometry (MFC-MRD) using a bone marrow aspirate, as standard-of-care. Overall, 57% MF-MRD tests were MRD(+) as defined by CLC numbers exceeding a threshold of 5 × 10-4%, which was determined to be the limit of quantitation. Above a threshold of 0.01%, MFC-MRD was positive in 34% of patients. The MF offered the advantage of the opportunity for efficiently processing small volumes of blood (2 mL), which is important in the care of pediatric patients, especially infants. The minimally invasive means of blood collection are of high value when treating patients whose MRD is typically tested using an invasive bone marrow biopsy. MF-MRD detection can be useful for stratification of patients into risk groups and monitoring of patient well-being after completion of treatment for early recognition of potential impending disease recurrence.
Collapse
Affiliation(s)
- Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA;
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
| | - Nicholas E. Larkey
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Alena Bartakova
- Biofluidica Inc., San Diego, CA 92121, USA; (A.B.); (M.L.H.)
| | | | - Shalee Mog
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
| | - Jami K. Cronin
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Judy Vun
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Keith J. August
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA;
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
- Biofluidica Inc., San Diego, CA 92121, USA; (A.B.); (M.L.H.)
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
3
|
Zala U, Patel R, Panchal V, Chaudhari J, Shah V, Shah A. Liquid biopsy for assessment of RFS (recurrence-free survival) in NSCLC (non-small cell lung cancer) patients post-treatment through circulating tumour DNA detection: A meta-analysis. THE JOURNAL OF LIQUID BIOPSY 2023; 2:100127. [PMID: 40028487 PMCID: PMC11863698 DOI: 10.1016/j.jlb.2023.100127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 03/05/2025]
Abstract
Background One of the main techniques for diagnosing lung cancer is still tissue biopsy. The more recent liquid biopsy technique includes the assessment of fragments of cfDNA, ctDNA, and CTCs, which are released by tumour cells and act as biomarkers. After compiling information from the existing literature, we conducted a statistical analysis to evaluate the prognosis and recurrence of NSCLC post-treatment using ctDNA levels. Method Based on predefined inclusion and exclusion criteria, published studies between 2000 and 2022 from electronic databases with statistical data on ctDNA levels for NSCLC were included. Then, using PRISMA guidelines, we carried out a systematic review of those studies. Result A total of 7 studies were included in the analysis, and we found a correlation of ctDNA levels for determination of prognosis and recurrence after treatment in NSCLC. Meta-analysis was performed using RevMan version 5.4. A P-value of less than 0.05 was considered significant. Studies were divided into three groups: <1 week post-surgery, one week to 3 months post-surgery, and > three months post-surgery. Analysis showed that in <1-week post-surgery group, ctDNA-negative post-treatment detection had an HR of 4.85 (95 % CI: 2.41 to 9.77; p=<0.0001, I2 = 0 %) for recurrence-free survival compared to ctDNA-positive post-treatment detection. Similar findings were obtained for one week to 3 months and > three months group, with HR of 5.27 (95 % CI: 3.62 to 7.67; p=<0.00001, I2 = 11 %) and HR of 6.62 (95 % CI: 2.85 to 15.35; p=<0.0001, I2 = 64 %) respectively. Conclusion According to our meta-analysis, patients with NSCLC who were ctDNA negative after surgery had a significantly longer recurrence-free survival than those who were ctDNA positive. As a result, liquid biopsy used to measure ctDNA levels is a useful non-invasive technique to assess the prognosis in such patients. The early detection of recurrence may also allow timely therapeutic intervention, leading to better outcomes.
Collapse
Affiliation(s)
- Urvi Zala
- Dept. of Medicine, Smt. N.H.L. Municipal Medical College and SVPISMR, Ahmedabad, Gujarat, India
| | - Rushi Patel
- Dept. of Medicine, Smt. N.H.L. Municipal Medical College and SVPISMR, Ahmedabad, Gujarat, India
| | - Viraj Panchal
- Dept. of Medicine, Smt. N.H.L. Municipal Medical College and SVPISMR, Ahmedabad, Gujarat, India
| | - Joy Chaudhari
- Dept. of Medicine, Smt. N.H.L. Municipal Medical College and SVPISMR, Ahmedabad, Gujarat, India
| | - Vedant Shah
- Dept. of Medicine, Smt. N.H.L. Municipal Medical College and SVPISMR, Ahmedabad, Gujarat, India
| | - Abhi Shah
- Dept. of Medicine, Smt. N.H.L. Municipal Medical College and SVPISMR, Ahmedabad, Gujarat, India
| |
Collapse
|
4
|
Shahkarami S, Younesian S, Rostami S, Kompani F, Bashash D, Vaezi M, Ghaffari SH. Minimal Residual Disease Detection Using Gene Scanning Analysis, Fluorescent Fragment Analysis, and Capillary Electrophoresis for IgH Rearrangement in Adult B-Lineage Acute Lymphoblastic Leukemia: A Cross-Sectional Study. CELL JOURNAL 2023; 25:85-91. [PMID: 36840454 PMCID: PMC9968371 DOI: 10.22074/cellj.2023.557390.1049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Indexed: 02/26/2023]
Abstract
OBJECTIVE Minimal residual disease (MRD) is considered the greatest prognostic factor in acute lymphoblastic leukemia (ALL). MRD is a valuable tool for anticipating impending relapse and treatment response assessment. The objective of the present study was to investigate whether the detection of IgH gene rearrangement using polymerase chain reaction (PCR)-based GeneScan analysis could be a complementary method to monitor MRD along with the quantitative realtime PCR (qPCR). MATERIALS AND METHODS In this cross-sectional study, we valued the MRD levels, based on the GeneScanning analysis (GSA), and then compared the data with quantitative real-time polymerase chain reaction at different time points in peripheral blood (PB) samples of adult B-lineage ALL patients (n=35). The specific polymerase chain reaction (PCR) primers for IGH gene FR-1 and fluorescence-labeled J-primer were used and analyzed by capillary gel electrophoresis on a sequencer. The results of this study were compared with the previously reported MRD results obtained by the IGH rearrangements allele-specific oligonucleotide (ASO) -qPCR methods. RESULTS The total concordance rate was 86.7%, with a P<0.001. MRD results obtained by GSA and ASO-qPCR methods were concordant in all diagnostic samples and samples on the 14th and 28th days of induction therapy. The results of these 2.5 years' follow-ups demonstrated a significant correlation between the two techniques (r=0.892, P<0.001). CONCLUSION It seems that the PCR-based GeneScan analysis of IGH gene rearrangement detection may be a valuable molecular technique to distinguish monoclonality from polyclonality. And also, it may be a precise tool to detect the residual leukemic DNA in the PB follow-up samples of patients.
Collapse
Affiliation(s)
- Sepideh Shahkarami
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical
Sciences, Tehran, Iran,Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, Ludwig-Maximilians-Universität München
(LMU), Munich, Germany
| | - Samareh Younesian
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical
Sciences, Tehran, Iran,Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical
Sciences, Tehran, Iran
| | - Shahrbano Rostami
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical
Sciences, Tehran, Iran
| | - Farzad Kompani
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical
Sciences, Tehran, Iran
| | - Mohammad Vaezi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical
Sciences, Tehran, Iran
| | - Seyed Hamidollah Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical
Sciences, Tehran, Iran,P.O.Box: 1411713135HematologyOncology and
Stem Cell Transplantation Research CenterShariati HospitalTehran University
of Medical SciencesTehranIran
| |
Collapse
|
5
|
Arabi S, Yousefian S, Kavosh A, Mansourian M, Nematollahi P. The prognostic significance of hematogones in childhood B-cell acute lymphoblastic leukemia. Pediatr Blood Cancer 2023; 70:e30138. [PMID: 36495254 DOI: 10.1002/pbc.30138] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Recent studies have demonstrated hematogones (HGs) expansion to be associated with favorable outcomes in hematological diseases, especially in patients with acute myeloid leukemia and patients undergoing hematopoietic stem cell transplantation. Acute lymphoblastic leukemia (ALL) is the most common form of cancer in children. As of now, minimal residual disease (MRD) remains the most compelling independent prognostic factor in childhood ALL. There is need for more prognostic tools for evaluating relapse risk. PROCEDURE The goal of this study was to assess the prognostic value of HGs on relapse-free survival (RFS) and overall survival (OS) in childhood ALL. In this prospective cohort study, a total of 122 subjects with definitive diagnosis of precursor B lymphoblastic leukemia were evaluated. Flow cytometric HG detection was performed in bone marrow aspirates after induction and consolidation therapy. RESULTS The median follow-up period of patients was 35.5 ± 9.4 (SD) months. Patients who had at least 1.0% HGs had a significantly better RFS (p = .023). Moreover, univariate and multivariate analyses confirmed that positive HGs were independently associated with longer RFS (unadjusted model: hazard ratio = 0.33, 95% CI = 0.12-0.91, p = .031; adjusted model: hazard ratio = 0.30, 95% CI = 0.11-0.82, p = .020). CONCLUSIONS Along with the role of MRD, our study shows the significance of HGs as an independent prognostic factor. The results indicate the independent prognostic value of HGs on RFS after adjustment for other prognostic factors, and can be beneficial for risk stratification and treatment modifications amongst pediatric B-cell ALL patients.
Collapse
Affiliation(s)
- Sina Arabi
- Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Yousefian
- Department of Pediatrics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aryan Kavosh
- Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marjan Mansourian
- Department of Epidemiology and Biostatistics, School of Public Health, Isfahan University of Medical Sciences, Isfahan, Iran.,Biomedical Engineering Research Centre (CREB), Automatic Control Department (ESAII), Universitat Politècnica de Catalunya-Barcelona, Barcelona, Spain
| | - Pardis Nematollahi
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Cancer Prevention Research Center, Omid Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
6
|
Thiotepa, busulfan and fludarabine conditioning-regimen is a promising approach for older adult patients with acute lymphoblastic leukemia treated with allogeneic stem cell transplantation. Bone Marrow Transplant 2023; 58:61-67. [PMID: 36224494 DOI: 10.1038/s41409-022-01841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 01/07/2023]
Abstract
For acute lymphoblastic leukemia (ALL) patients, total body irradiation (TBI)- based conditioning regimens are the first choice specially in young population. However, several studies have shown an equivalence in clinical outcomes with thiotepa-based conditioning regimen. We performed a retrospective study to evaluate the outcome of adult ALL patients who received allogeneic hematopoietic stem cell transplantation (allo-HCT) with a thiotepa-busulfan-fludarabine (TBF) myeloablative conditioning regimen with reduced toxicity. Fifty-five patients received a TBF regimen. The median age of the patients was 51 years (range, 17 to 72.4). Most patients had a diagnosis of B-ALL (93%) with 7% having T-ALL. Two - and 5-year overall survival was 73.2% and 64%, respectively. At 2 years, leukemia-free survival and GVHD-free, relapse-free survival were 59.5% and 57.6%, and at 5 years, 53.4% and 51.8%, respectively. The 5-year non-relapse mortality was 15%. The day 180 cumulative incidence (CI) of grade II-IV acute GVHD and grade III-IV acute GVHD were 38.2% and 5.5%, respectively. At 2 years, the CI of chronic GVHD and extensive chronic GVHD was 16.9% and 1.9%, respectively. Our study results do suggest that using TBF as the conditioning regimen in adult ALL patients is a promising option with acceptable toxicity.
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW While the treatment of acute lymphoblastic leukemia (ALL) has improved significantly over the last 30 years, the majority of adult patients will have their disease relapse. The BCL-2 gene was initially discovered from follicular lymphoma research; however, the BH3 family of proteins has is emerging to be crucial in patients with ALL due to their reliance on the balance of these pro-apoptotic and anti-apoptotic proteins in the BH3 family. We discuss apoptosis in ALL, the reliance mechanisms, drug development in this space, and areas for future research. RECENT FINDINGS The first drugs that were developed to inhibit the BCL-2 pathway include both venetoclax (BCL-2 specific inhibitor) and navitoclax (BCL-2, BCL-XL, and BCL-W). These drugs show promise and have obtained complete remissions, minimal residual disease negative status, and have been used as a bridge to allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia and chronic lymphocytic leukemia. There are multiple ongoing clinical trials looking to assess the use of BCL-2 inhibition with chemotherapy, targeted therapies, and bi-specific T-cell engager therapies not only in both frontline and relapsed refractory ALL but also in consolidation and maintenance phases. There is still a large need for improvement of ALL outcomes in adult patients. Research has shown that ALL depends on the BCL-2 family of proteins for cell survival and proliferation. Targeting this pathway with BCL-2 inhibition has led to encouraging results, and future research is aimed at incorporating this targeted therapy into current treatment paradigms.
Collapse
Affiliation(s)
- Wesley M Smith
- Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, USA
| | - Daniel R Reed
- Comprehensive Cancer Center of Wake Forest University, Winston Salem, NC, USA.
| |
Collapse
|
8
|
Liang EC, Craig J, Torelli S, Cunanan K, Iglesias M, Arai S, Frank MJ, Johnston L, Lowsky R, Meyer EH, Miklos DB, Negrin R, Rezvani A, Shiraz P, Shizuru J, Sidana S, Weng WK, Bharadwaj S, Muffly L. Allogeneic Hematopoietic Cell Transplantation for Adult Acute Lymphoblastic Leukemia in the Modern Era. Transplant Cell Ther 2022; 28:490-495. [PMID: 35584783 PMCID: PMC10153066 DOI: 10.1016/j.jtct.2022.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/19/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) remains an important treatment for adults with acute lymphoblastic leukemia (ALL). We hypothesized that advances in ALL and transplantation have resulted in improved HCT outcomes in recent years. In this study, we evaluated the characteristics and outcomes of adult ALL patients undergoing allogeneic HCT over the last decade. Patients with ALL aged 18 years and older who underwent allogeneic HCT at Stanford University between 2008 and 2019 were included in this study. Patients were divided into 2 eras based on year of HCT: 2008 to 2013 (earlier era) and 2014 to 2019 (later era). A total of 285 patients were included: 119 patients underwent HCT in the earlier era and 166 in the later era. Patients who underwent transplantation in the later era were more likely to be Hispanic (38% versus 21%) and to have an HCT-comorbidity index ≥3 (31% versus 18%). Donor source for HCT also differed with an increase in the use of HLA-mismatched donor sources (38% versus 24%), notably umbilical cord blood in the later era (16% versus 0%). Patients in the later era were less likely to undergo transplantation with active disease (4% versus 16%); pre-HCT rates of measurable residual disease were similar across the eras (38% versus 40%). In unadjusted analyses, overall survival (OS) improved across eras, with 2-year estimates for the later and earlier eras of 73% (95% confidence interval [CI], 66%-80%) versus 55% (95% CI, 46%-64%), respectively. Multivariable analysis confirmed the association between later era and OS (hazard ratio = 0.52, 95% CI, 0.34-0.78). Finally, among patients relapsing after HCT (25% in later era and 33% in earlier era), the use of novel immunotherapies increased in the later era (44% versus 3%), as did the median OS after post-HCT relapse (16 months versus 8 months, P< .001). OS after HCT for adult ALL has improved in recent years. This is due, in part, to a significant improvement in the ability to effectively salvage adults with ALL relapsing after HCT.
Collapse
Affiliation(s)
- Emily C Liang
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Juliana Craig
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Stefan Torelli
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kristen Cunanan
- Quantitative Sciences Unit, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Maria Iglesias
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Sally Arai
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Matthew J Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Laura Johnston
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Robert Lowsky
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Everett H Meyer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - David B Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Robert Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Andrew Rezvani
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Parveen Shiraz
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Judith Shizuru
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Sushma Bharadwaj
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
9
|
Toksvang LN, Grell K, Nersting J, Degn M, Nielsen SN, Abrahamsson J, Lund B, Kanerva J, Jónsson ÓG, Lepik K, Vaitkevičienė G, Griškevičius L, Quist-Paulsen P, Vora A, Moorman AV, Murdy D, Zimmermann M, Möricke A, Bostrom B, Joshi J, Hjalgrim LL, Dalhoff KP, Als-Nielsen B, Schmiegelow K. DNA-thioguanine concentration and relapse risk in children and young adults with acute lymphoblastic leukemia: an IPD meta-analysis. Leukemia 2022; 36:33-41. [PMID: 34175901 DOI: 10.1038/s41375-021-01182-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/11/2021] [Accepted: 02/02/2021] [Indexed: 12/17/2022]
Abstract
Methotrexate/6-mercaptopurine maintenance therapy improves acute lymphoblastic leukemia (ALL) outcome. Cytotoxicity is mediated by DNA incorporation of thioguanine nucleotides (DNA-TG). We investigated the association of DNA-TG to relapse risk in 1 910 children and young adults with non-high risk ALL. In a cohort-stratified Cox regression analysis adjusted for sex, age, and white cell count at diagnosis, the relapse-specific hazard ratio (HRa) per 100 fmol/μg increase in weighted mean DNA-TG (wmDNA-TG) was 0.87 (95% CI 0.78-0.97; p = 0.013) in the 839 patients who were minimal residual disease (MRD) positive at end of induction therapy (EOI), whereas this was not the case in EOI MRD-negative patients (p = 0.76). Validation analysis excluding the previously published Nordic NOPHO ALL2008 pediatric cohort yielded a HRa of 0.92 (95% CI 0.82-1.03; p = 0.15) per 100 fmol/μg increase in wmDNA-TG in EOI MRD-positive patients. If also excluding the United Kingdom cohort, in which samples were taken non-randomly in selected patients, the HRa for the EOI MRD-positive patients was 0.82 (95% CI 0.68-0.99; p = 0.044) per 100 fmol/μg increase in wmDNA-TG. The importance of DNA-TG as a biomarker for maintenance therapy intensity calls for novel strategies to increase DNA-TG, although its clinical value may vary by protocol backbone.
Collapse
Affiliation(s)
| | - Kathrine Grell
- Rigshospitalet, Copenhagen, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | - Bendik Lund
- St Olavs Hospital, Trondheim, Norway
- Norwegian University of Science and Technology, Trondheim, Norway
| | - Jukka Kanerva
- Helsinki Central Hospital and University of Helsinki, Helsinki, Finland
| | | | | | - Goda Vaitkevičienė
- Center for Pediatric Oncology and Hematology, Vilnius University, Vilnius, Lithuania
| | - Laimonas Griškevičius
- Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Vilnius University, Vilnius, Lithuania
| | | | - Ajay Vora
- Great Ormond Street Hospital for Children National Health Service Trust, London, United Kingdom
| | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Daniel Murdy
- Leukaemia Research Cytogenetics Group, Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | | | - Anja Möricke
- University Hospital Schleswig-Holstein, Kiel, Germany
| | - Bruce Bostrom
- Children's Hospital and Clinics of Minnesota, Minneapolis, MI, USA
| | - Jaitri Joshi
- Children's Hospital and Clinics of Minnesota, Minneapolis, MI, USA
| | | | - Kim P Dalhoff
- University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg University Hospital, Copenhagen, Denmark
| | | | - Kjeld Schmiegelow
- Rigshospitalet, Copenhagen, Denmark.
- University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
10
|
Fernando F, Robertson HF, El-Zahab S, Pavlů J. How I Use Measurable Residual Disease in the Clinical Management of Adult Acute Lymphoblastic Leukemia. Clin Hematol Int 2021; 3:130-141. [PMID: 34938985 PMCID: PMC8690704 DOI: 10.2991/chi.k.211119.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/05/2021] [Indexed: 11/01/2022] Open
Abstract
Over the last decade the use of measurable residual disease (MRD) diagnostics in adult acute lymphoblastic leukemia (ALL) has expanded from a limited number of study groups in Europe and the United States to a world-wide application. In this review, we summarize the advantages and drawbacks of the current available techniques used for MRD monitoring. Through the use of three representative case studies, we highlight the advances in the use of MRD in clinical decision-making in the management of ALL in adults. We acknowledge discrepancies in MRD monitoring and treatment between different countries, reflecting differing availability, accessibility and affordability.
Collapse
Affiliation(s)
- Fiona Fernando
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | | | - Sarah El-Zahab
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Jiří Pavlů
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| |
Collapse
|
11
|
Saleh M, Khalil M, Abdellateif MS, Ebeid E, Madney Y, Kandeel EZ. Role of matrix metalloproteinase MMP-2, MMP-9 and tissue inhibitor of metalloproteinase (TIMP-1) in the clinical progression of pediatric acute lymphoblastic leukemia. ACTA ACUST UNITED AC 2021; 26:758-768. [PMID: 34555302 DOI: 10.1080/16078454.2021.1978763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) play a crucial role in cancer progression and metastasis, however their role in pediatric Acute lymphoblastic leukemia (ALL) is still unrevealed. METHODS The diagnostic, prognostic and predictive value of tissue inhibitor of metalloproteinase (TIMP-1), MMP-2, MMP-9 and CD34+CD38- cancer stem cells (CSCs) were assessed in bone marrow (BM) samples of 76 ALL children using Flow Cytometry analysis. RESULTS There was a significant increase in TIMP-1 [1.52 (0.41-10) versus 0.91(0.6-1.12); respectively, p < 0.001], and CSCs CD34+CD38- [1 (0.03-18.6) versus 0.3 (0.01-1.1), p < 0.001] expression in ALL patients compared to controls. While there were no significant differences regarding MMP-2 and MMP-9 expression between the two groups. The sensitivity, specificity, area under curve (AUC) of MMP-2 were (80.3%, 53.3% and 0.568, p = 0.404), and of MMP-9 were (53.9%, 40% and 0.660, p = 0.053). While that of TIMP-1 were (78.9%, 100% and 0.892, p < 0.001), and that of CD34+CD38- CSCs were (78.9%, 73.3% and 0.855, p < 0.001). Increased TIMP-1 expression associated with the high-risk disease (p < 0.001). CD34+CD38- CSCs and MMP-2 overexpression associated with MRD at day-15, increased BM blast cell count at diagnosis and at day-15 (p < 0.05). TIMP-1 overexpression is associated with shorter DFS and OS rates (p = 0.009 and p = 0.048). Multivariate logistic regression analysis showed that both TIMP-1 [OR: 4.224, p = 0.046], and CD34+CD38- CSCs [OR: 6.873, p = 0.005] could be potential independent diagnostic factors for pediatric ALL. CONCLUSION TIMP-1 and CD34+CD38- CSCs could be possible useful diagnostic markers for pediatric ALL. Also, TIMP-1 is a promising prognostic marker for poor outcome of the patients.
Collapse
Affiliation(s)
- Maha Saleh
- Clinical Pathology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Mohamed Khalil
- Clinical Pathology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Emad Ebeid
- Pediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Youssef Madney
- Pediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Eman Z Kandeel
- Clinical Pathology, National Cancer Institute, Cairo University, Giza, Egypt
| |
Collapse
|
12
|
Virk H, Rana S, Sharma P, Bose PL, Yadav DD, Sachdeva MUS, Varma N, Trehan A, Lad D, Khadwal AR, Malhotra P, Sreedharanunni S. Hematological characteristics, cytogenetic features, and post-induction measurable residual disease in thymic stromal lymphopoietin receptor (TSLPR) overexpressed B-cell acute lymphoblastic leukemia in an Indian cohort. Ann Hematol 2021; 100:2031-2041. [PMID: 34159401 DOI: 10.1007/s00277-021-04574-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/08/2021] [Indexed: 11/24/2022]
Abstract
The overexpression of cytokine receptor-like factor-2 (CRLF2) identified by anti-thymic stromal lymphopoietin receptor/TSLPR flow cytometry (FCM) has been reported as a screening tool for the identification of BCR-ABL1-like B-cell acute lymphoblastic leukemia/B-ALL with CRLF2 re-arrangement. TSLPR expression was studied prospectively in consecutive 478 B-ALLs (≤ 12 years (n = 244); 13-25 years (n = 129); > 25 years (n = 105)) and correlated with various hematological parameters and end-of-induction measurable residual disease (day 29; MRD ≥ 0.01% by 10-color FCM). TSLPR positivity in ≥ 10% leukemic cells was detected in 14.6% (n = 70) of B-ALLs. CRLF2 re-arrangement was detected in eight cases (11.4%) including P2RY8-CRLF2 (n = 6), and IgH-CRLF2 (n = 2) with a median TSLPR positivity of 48.8% and 99% leukemic cells, respectively. Recurrent gene fusions/RGF (BCR-ABL1 (17.1%); ETV6-RUNX1 (4.2%), TCF3-PBX1 (1.4%)), other BCR-ABL1-like chimeric gene fusions/CGFs (PDGFRB-rearrangement (2.9%), IgH-EPOR (1.4%)), CRLF2 extra-copies/hyperdiploidy (17.1%), and IgH translocation without a known partner (10%) were also detected in TSLPR-positive patients. CD20 positivity (52.9% vs 38.5%; p = 0.02) as well as iAMP21 (4.3% vs 0.5%; p = 0.004) was significantly more frequent in TSLPR-positive cases. TSLPR-positive patients did not show a significantly higher MRD, compared to TSLPR-negative cases (37% vs 33%). Increasing the threshold cut-off (from ≥ 10 to > 50% or > 74%) increased the specificity to 88% and 100% respectively in identifying CRLF2 translocation. TSLPR expression is not exclusive for CRLF2 translocations and can be seen with various other RGFs, necessitating their testing before its application in diagnostic algorithms. In patients with high TSLPR positivity (> 50%), the testing may be restricted to CRLF2 aberrancies, while patients with 10-50% TSLPR positivity need to be tested for both CRLF2- and non-CRLF2 BCR-ABL1-like CGFs.
Collapse
Affiliation(s)
- Harpreet Virk
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India, 160012
| | - Sonia Rana
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India, 160012
| | - Praveen Sharma
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India, 160012
| | - Parveen Lata Bose
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India, 160012
| | - Diksha Dev Yadav
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India, 160012
| | - Man Updesh Singh Sachdeva
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India, 160012
| | - Neelam Varma
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India, 160012
| | - Amita Trehan
- Pediatric Hematology/Oncology Unit, Department of Pediatric Medicine, Advanced Pediatric Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepesh Lad
- Adult Clinical Hematology Unit, Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Rani Khadwal
- Adult Clinical Hematology Unit, Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Adult Clinical Hematology Unit, Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sreejesh Sreedharanunni
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India, 160012.
| |
Collapse
|
13
|
Kantarjian HM, Stock W, Cassaday RD, DeAngelo DJ, Jabbour E, O'Brien SM, Stelljes M, Wang T, Paccagnella ML, Nguyen K, Sleight B, Vandendries E, Neuhof A, Laird AD, Advani AS. Inotuzumab Ozogamicin for Relapsed/Refractory Acute Lymphoblastic Leukemia in the INO-VATE Trial: CD22 Pharmacodynamics, Efficacy, and Safety by Baseline CD22. Clin Cancer Res 2021; 27:2742-2754. [PMID: 33602684 DOI: 10.1158/1078-0432.ccr-20-2399] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 12/03/2020] [Accepted: 02/10/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE We assessed the relationship between cluster of differentiation-22 (CD22) expression and outcomes of inotuzumab ozogamicin versus standard of care (SC) in INO-VATE (NCT01564784). PATIENTS AND METHODS Adults with relapsed/refractory B-cell precursor CD22-positive (by local or central laboratory) acute lymphoblastic leukemia were randomized to inotuzumab ozogamicin (n = 164) or SC (n = 162). Outcomes were analyzed by baseline CD22 positivity (percentage of leukemic blasts CD22 positive, ≥90% vs. <90%) and CD22 receptor density [molecules of equivalent soluble fluorochrome (MESF), quartile analysis]. RESULTS Most patients had high (≥90%) CD22 positivity per central laboratory. The response rate was significantly higher with inotuzumab ozogamicin versus SC. Minimal/measurable residual disease negativity, duration of remission (DoR), progression-free survival, and overall survival (OS) were significantly better with inotuzumab ozogamicin versus SC in patients with CD22 positivity ≥90%. Fewer patients had CD22 positivity <90%; for whom, response rates were higher with inotuzumab ozogamicin versus SC, but DoR and OS appeared similar. Similar trends were evident in quartile analyses of CD22 MESF and CD22 positivity per local laboratory. Among inotuzumab ozogamicin-responding patients with subsequent relapse, decrease in CD22 positivity and receptor density was evident, but not the emergence of CD22 negativity. Rates of grade ≥3 hematologic adverse events (AEs) were similar and hepatobiliary AEs rate was higher for inotuzumab ozogamicin versus SC. No apparent relationship was observed between the rates of hematologic and hepatic AEs and CD22 expression. CONCLUSIONS Inotuzumab ozogamicin demonstrated a favorable benefit-risk profile versus SC in patients with higher and lower CD22 expression. Patients with high CD22 expression and normal cytogenetics benefited the most from inotuzumab ozogamicin therapy.
Collapse
Affiliation(s)
| | | | - Ryan D Cassaday
- University of Washington School of Medicine and Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Elias Jabbour
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Susan M O'Brien
- Chao Family Comprehensive Cancer Center, University of California Irvine, Orange, California
| | | | - Tao Wang
- Pfizer Inc., Cambridge, Massachusetts
| | | | - Kevin Nguyen
- Navigate BioPharma Services, Inc., a Novartis subsidiary, Carlsbad, California
| | | | | | | | | | | |
Collapse
|
14
|
Hassan NM, Refaat LA, Ismail GN, Abdellateif M, Fadel SA, AbdelAziz RS. Diagnostic, prognostic and predictive values of miR-100 and miR-210 in pediatric acute lymphoblastic Leukemia. ACTA ACUST UNITED AC 2021; 25:405-413. [PMID: 33191875 DOI: 10.1080/16078454.2020.1843753] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND : microRNAs are playing important roles in the diagnosis and prognosis of pediatric acute lymphoblastic leukemia (ALL). METHODS Expression levels of miR-100 and miR-210 were assessed in bone marrow aspirate of 85 pediatric ALL patients compared to 12 healthy control using quantitative real-time polymerase chain reaction. Data were correlated with relevant clinico-pathological features of the patients, response to treatment, disease-free survival (DFS), and overall survival (OS). RESULTS miR-100 was significantly downregulated in ALL patients [median: 1.21, range: 0-434.3] compared to the control group [median: 8.41, range; 0-840.3, P = 0.035]. miR-210 was significantly upregulated in ALL patients [median: 6.34, range: 1.16-1088.7] compared to the control group [median: 2.57, range: 0.11-709.2, P = 0.025]. The sensitivity, specificity, and area under curve of miR-100 were (64.7%, 62.5%, and 0.642; respectively, P = 0.035) at a cut-off 2.6 and that of miR-210 were (60%, 58.3% and 0.650; respectively, P = 0.025) at a cut-off 3.5. miR-100 overexpression associated with shorter DFS and OS (P = 0.033 and 0.046; respectively). Patients with miR-100 lowexpression showed a significant incidence of late death ( P = 0.024). There was no significant association between miR-210 expression and DFS, OS, incidence of early or late death. CONCLUSION : miR-100 and miR-210 could be used as potential diagnostic markers for pediatric ALL. miR-100 is a useful prognostic and predictive biomarker for childhood ALL.
Collapse
Affiliation(s)
- Naglaa M Hassan
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Lobna A Refaat
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Ghada N Ismail
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mona Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Sayed A Fadel
- Pediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Rania S AbdelAziz
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
15
|
Halford Z, Coalter C, Gresham V, Brown T. A Systematic Review of Blinatumomab in the Treatment of Acute Lymphoblastic Leukemia: Engaging an Old Problem With New Solutions. Ann Pharmacother 2021; 55:1236-1253. [PMID: 33435716 DOI: 10.1177/1060028020988411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the current literature for blinatumomab in the treatment of adult and pediatric B-cell acute lymphoblastic leukemia (ALL). DATA SOURCES We conducted a PubMed (inception to December 11, 2020) and ClinicalTrials.gov systematic literature search using the following terms: blinatumomab, Blincyto, lymphoblastic leukemia, and bispecific T-cell engager. STUDY SELECTION AND DATA EXTRACTION All relevant published articles, package inserts, and meeting abstracts evaluating the use of blinatumomab in ALL were considered for inclusion. DATA SYNTHESIS Blinatumomab, a first-in-class bispecific T-cell engager monoclonal antibody, facilitates cytotoxic T-cell activation and subsequent eradication of CD19-positive B cells. The confirmatory phase III TOWER trial demonstrated superior overall survival (OS) with blinatumomab compared with standard chemotherapy (7.7 months vs 4.0 months) in relapsed and refractory (R/R) B-cell ALL. In the phase II BLAST trial, blinatumomab achieved a complete measurable residual disease (MRD) response in 78% of evaluable patients, with a median OS of 36.5 months. Potentially life-threatening cytokine release syndrome and neurotoxicity occurred in approximately 15% and 65% of patients, respectively. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Following initial Food and Drug Administration approval in 2014, blinatumomab gained expanded approval in pediatric patients and in Philadelphia chromosome-positive R/R ALL. In 2018, blinatumomab became the first and only drug approved for the treatment of persistent MRD in any hematologic malignancy. Emerging data demonstrate promising efficacy with blinatumomab in specific ALL settings, including frontline therapy, as a bridge to transplantation, and in "chemotherapy-free" combination regimens. CONCLUSIONS Blinatumomab provides a paradigm-shifting treatment option; however, many questions surrounding optimal patient selection, sequencing, and cost-effectiveness remain.
Collapse
Affiliation(s)
| | - Carli Coalter
- Union University College of Pharmacy, Jackson, TN, USA
| | | | - Tabitha Brown
- Erlanger Health System/Children's Hospital at Erlanger, Chattanooga, TN, USA
| |
Collapse
|
16
|
Correia RP, Puga RD, Muto NH, Lee MLDM, Torres DC, Hassan R, Bacal NS, Hamerschlak N, Campregher PV. High-throughput sequencing of immunoglobulin heavy chain for minimal residual disease detection in B-lymphoblastic leukemia. Int J Lab Hematol 2021; 43:724-731. [PMID: 33393719 DOI: 10.1111/ijlh.13453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/24/2020] [Accepted: 12/17/2020] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Minimal residual disease (MRD) is a cornerstone for stratification of upfront B-lymphoblastic leukemia (B-ALL) treatment protocols to decrease relapse risk. Although its detection by flow cytometry (FC) and real-time quantitative polymerase has clinical usefulness, evidence suggests that methods with increased sensitivity could lead to improved outcomes. The aim of this study was to develop an amplicon-based assay followed by high-throughput sequencing of the immunoglobulin heavy chain variable region for MRD detection in B-ALL. METHODS We analyzed 84 samples, 27 from diagnosis, 5 from relapse, 40 from post-treatment samples, and 12 from healthy controls. RESULTS Our assay was able to identify more neoplastic clones at diagnosis than Sanger sequencing including incomplete DJ rearrangements. From the 40 MRD samples evaluated 21 were positive by our new approach on high-throughput sequencing assay, but only 15 of these were positive by FC. The remaining 19 were negative by the two techniques. CONCLUSION We have developed a novel approach on high-sensitive assay for MRD detection in B-ALL, which could add clinical value in the management of patients, especially in cases negative for MRD by FC.
Collapse
Affiliation(s)
- Rodolfo P Correia
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Renato D Puga
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Nair H Muto
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Davi C Torres
- Bone Marrow Transplantation Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Rocio Hassan
- Bone Marrow Transplantation Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Nydia S Bacal
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Centro de Hematologia de São Paulo, São Paulo, Brazil
| | - Nelson Hamerschlak
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Paulo V Campregher
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
17
|
Agarwal M, Seth R, Chatterjee T. Recent Advances in Molecular Diagnosis and Prognosis of Childhood B Cell Lineage Acute Lymphoblastic Leukemia (B-ALL). Indian J Hematol Blood Transfus 2021; 37:10-20. [PMID: 33707831 PMCID: PMC7900311 DOI: 10.1007/s12288-020-01295-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/25/2020] [Indexed: 11/26/2022] Open
Abstract
B cell lineage acute lymphoblastic leukemia is the most common leukemia occurring in children and young adults and is the leading cause of cancer related deaths. The 5 year overall survival outcome in children with B-ALL has improved significantly in the last few decades. In the past, the discovery of various genetic alterations and targeted therapy have played a major role in decreasing disease-related deaths. In addition, numerous advances in the pathogenesis of B-ALL have been found which have provided better understanding of the genes involved in disease biology with respect to diagnostic and prognostic implications. Present review will summarize current understanding of risk stratification, genetic factors including cytogenetics in diagnosis and prognosis of B-ALL.
Collapse
Affiliation(s)
- Manisha Agarwal
- Department of Laboratory Sciences and Molecular Medicine, Army Hospital (R&R), New Delhi, India
| | - Rachna Seth
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Tathagata Chatterjee
- Department of Laboratory Sciences and Molecular Medicine, Army Hospital (R&R), New Delhi, India
| |
Collapse
|
18
|
Fox TA, Carpenter B, Taj M, Perisoglou M, Nicholson E, Castleton A, Elliot J, Uttenthal B, Wright C, Halsey R, Khwaja A, Grandage V, Mansour MR, Fielding AK, Hough R. Utility of 18F-FDG-PET/CT in lymphoblastic lymphoma. Leuk Lymphoma 2020; 62:1010-1012. [PMID: 33275056 DOI: 10.1080/10428194.2020.1855346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Thomas A Fox
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Ben Carpenter
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Mary Taj
- Department of Clinical Haematology, Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Martha Perisoglou
- Department of Clinical Haematology, Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Emma Nicholson
- Department of Clinical Haematology, Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Anna Castleton
- Department of Clinical Haematology, The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Johnathon Elliot
- Department of Clinical Haematology, The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Ben Uttenthal
- Department of Clinical Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Callum Wright
- Department of Clinical Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Richard Halsey
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - Asim Khwaja
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, UK.,UCL Cancer Institute, University College London, London, UK
| | - Victoria Grandage
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Marc R Mansour
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, UK.,UCL Cancer Institute, University College London, London, UK
| | - Adele K Fielding
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, UK.,UCL Cancer Institute, University College London, London, UK
| | - Rachael Hough
- Department of Clinical Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
19
|
Greil C, Engelhardt M, Ihorst G, Duque-Afonso J, Shoumariyeh K, Bertz H, Marks R, Zeiser R, Duyster J, Finke J, Wäsch R. Prognostic factors for survival after allogeneic transplantation in acute lymphoblastic leukemia. Bone Marrow Transplant 2020; 56:841-852. [PMID: 33130821 PMCID: PMC8266681 DOI: 10.1038/s41409-020-01101-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/23/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Allogeneic stem cell transplantation (allo-SCT) offers a curative option in adult patients with acute lymphoblastic leukemia (ALL). Prognostic factors for survival after allo-SCT have not been sufficiently defined: pheno-/genotype, patients´ age, conditioning regimens and remission at allo-SCT are under discussion. We analyzed the outcome of 180 consecutive adult ALL-patients undergoing allo-SCT at our center between 1995 and 2018 to identify specific prognostic factors. In our cohort 19% were older than 55 years, 28% had Philadelphia-positive B-ALL, 24% T-ALL. 54% were transplanted in first complete remission (CR1), 13% in CR2 after salvage therapy, 31% reached no remission (8% within first-line, 23% within salvage therapy). In 66% conditioning contained total body irradiation (TBI). With a median follow-up of 10 years, we observed an overall survival of 33% at 10 years, and a progression free survival of 31%. The cumulative incidence of relapse was 41% at 10 years, the cumulative incidence of non-relapse mortality 28%. Acute graft-versus-host disease (GvHD) II°-IV° occurred in 31%, moderate/severe chronic GvHD in 27%. Survival was better in patients reaching CR before allo-SCT and in those receiving TBI. No difference between patients younger/older than 55 years and between different phenotypes was observed. Survival after allo-SCT improved considerably over the last decades.
Collapse
Affiliation(s)
- C Greil
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - M Engelhardt
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - G Ihorst
- Clinical Trials Unit, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - J Duque-Afonso
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - K Shoumariyeh
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - H Bertz
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - R Marks
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - R Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - J Duyster
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - J Finke
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - R Wäsch
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany. .,Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
20
|
Microfluidic Chip based direct triple antibody immunoassay for monitoring patient comparative response to leukemia treatment. Biomed Microdevices 2020; 22:48. [PMID: 32661698 DOI: 10.1007/s10544-020-00503-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We report a time and cost-efficient microfluidic chip for screening the leukemia cells having three specific antigens. In this method, the target blast cells are double sorted with immunomagnetic beads and captured by the 3rd antibody immobilized on the gold surface in a microfluidic chip. The captured blast cells in the chip were imaged using a bright-field optical microscope and images were analyzed to quantify the cells. First sorting was performed with nano size immunomagnetic beads and followed by 2nd sorting where micron size immunomagnetic beads were used. The low-cost microfluidic platform is made of PMMA and glass including micro size gold pads. The developed microfluidic platform was optimized with cultured B type lymphoblast cells and tested with the samples of leukemia patients. The 8 bone marrow samples of 4 leukemia patients on the initial diagnosis and on the 15th day after the start of the chemotherapy treatment were tested both with the developed microfluidic platform and the flow cytometry. A 99% statistical agreement between the two methods shows that the microfluidic chip is able to monitor the decrease in the number of blast cells due to the chemotherapy. The experiments with the patient samples demonstrate that the developed system can perform relative measurements and have a potential to monitor the patient response to the applied therapy and to enable personalized dose adjustment.
Collapse
|
21
|
Autry RJ, Paugh SW, Carter R, Shi L, Liu J, Ferguson DC, Lau CE, Bonten EJ, Yang W, McCorkle JR, Beard JA, Panetta JC, Diedrich JD, Crews KR, Pei D, Coke CJ, Natarajan S, Khatamian A, Karol SE, Lopez-Lopez E, Diouf B, Smith C, Gocho Y, Hagiwara K, Roberts KG, Pounds S, Kornblau SM, Stock W, Paietta EM, Litzow MR, Inaba H, Mullighan CG, Jeha S, Pui CH, Cheng C, Savic D, Yu J, Gawad C, Relling MV, Yang JJ, Evans WE. Integrative genomic analyses reveal mechanisms of glucocorticoid resistance in acute lymphoblastic leukemia. NATURE CANCER 2020; 1:329-344. [PMID: 32885175 PMCID: PMC7467080 DOI: 10.1038/s43018-020-0037-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 01/29/2020] [Indexed: 12/31/2022]
Abstract
Identification of genomic and epigenomic determinants of drug resistance provides important insights for improving cancer treatment. Using agnostic genome-wide interrogation of mRNA and miRNA expression, DNA methylation, SNPs, CNAs and SNVs/Indels in primary human acute lymphoblastic leukemia cells, we identified 463 genomic features associated with glucocorticoid resistance. Gene-level aggregation identified 118 overlapping genes, 15 of which were confirmed by genome-wide CRISPR screen. Collectively, this identified 30 of 38 (79%) known glucocorticoid-resistance genes/miRNAs and all 38 known resistance pathways, while revealing 14 genes not previously associated with glucocorticoid-resistance. Single cell RNAseq and network-based transcriptomic modelling corroborated the top previously undiscovered gene, CELSR2. Manipulation of CELSR2 recapitulated glucocorticoid resistance in human leukemia cell lines and revealed a synergistic drug combination (prednisolone and venetoclax) that mitigated resistance in mouse xenograft models. These findings illustrate the power of an integrative genomic strategy for elucidating genes and pathways conferring drug resistance in cancer cells.
Collapse
Affiliation(s)
- Robert J Autry
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Steven W Paugh
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Robert Carter
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lei Shi
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jingjing Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Daniel C Ferguson
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Calvin E Lau
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
- Pediatric Oncology Education Program, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Erik J Bonten
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wenjian Yang
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - J Robert McCorkle
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jordan A Beard
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - John C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jonathan D Diedrich
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kristine R Crews
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Deqing Pei
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Christopher J Coke
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sivaraman Natarajan
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alireza Khatamian
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Seth E Karol
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Elixabet Lopez-Lopez
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Barthelemy Diouf
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Colton Smith
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yoshihiro Gocho
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kohei Hagiwara
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kathryn G Roberts
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Steven M Kornblau
- Department of Leukemia, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy Stock
- Hematopoiesis and Hematological Malignancies Program, University of Chicago, Chicago, IL, USA
| | - Elisabeth M Paietta
- Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, North Division, Bronx, NY, USA
| | - Mark R Litzow
- Division of Hematology and Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hiroto Inaba
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Charles G Mullighan
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sima Jeha
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Daniel Savic
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Charles Gawad
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mary V Relling
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jun J Yang
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - William E Evans
- Hematological Malignancies Program and Center for Precision Medicine in Leukemia, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
22
|
Liu HC, Gang EJ, Kim HN, Lim HG, Jung H, Chen R, Abdel-Azim H, Shung KK, Kim YM. Characterizing Deformability of Drug Resistant Patient-Derived Acute Lymphoblastic Leukemia (ALL) Cells Using Acoustic Tweezers. Sci Rep 2018; 8:15708. [PMID: 30356155 PMCID: PMC6200731 DOI: 10.1038/s41598-018-34024-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 10/05/2018] [Indexed: 12/31/2022] Open
Abstract
The role of cell mechanics in cancer cells is a novel research area that has resulted in the identification of new mechanisms of therapy resistance. Single beam acoustic (SBA) tweezers are a promising technology for the quantification of the mechanical phenotype of cells. Our previous study showed that SBA tweezers can be used to quantify the deformability of adherent breast cancer cell lines. The physical properties of patient-derived (primary) pre-B acute lymphoblastic leukemia (ALL) cells involved in chemotherapeutic resistance have not been widely investigated. Here, we demonstrate the feasibility of analyzing primary pre-B ALL cells from four cases using SBA tweezers. ALL cells showed increased deformability with increasing acoustic pressure of the SBA tweezers. Moreover, ALL cells that are resistant to chemotherapeutic drugs were more deformable than were untreated ALL cells. We demonstrated that SBA tweezers can quantify the deformability of nonadherent leukemia cells and discriminate this mechanical phenotype in chemotherapy-resistant leukemia cells in a contact- and label-free manner.
Collapse
Affiliation(s)
- Hsiao-Chuan Liu
- Department of Biomedical Engineering and NIH Ultrasonic Transducer Resource Center, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA.,Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA, 90027, USA
| | - Eun Ji Gang
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA, 90027, USA
| | - Hye Na Kim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA, 90027, USA
| | - Hae Gyun Lim
- Department of Creative IT Engineering and Future IT Innovation Laboratory, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Hayong Jung
- Department of Biomedical Engineering and NIH Ultrasonic Transducer Resource Center, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA
| | - Ruimin Chen
- Department of Biomedical Engineering and NIH Ultrasonic Transducer Resource Center, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA
| | - Hisham Abdel-Azim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA, 90027, USA
| | - K Kirk Shung
- Department of Biomedical Engineering and NIH Ultrasonic Transducer Resource Center, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA.
| | - Yong-Mi Kim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA, 90027, USA.
| |
Collapse
|
23
|
Weston VJ, Wei W, Stankovic T, Kearns P. Synergistic action of dual IGF1/R and MEK inhibition sensitizes childhood acute lymphoblastic leukemia (ALL) cells to cytotoxic agents and involves downregulation of STAT6 and PDAP1. Exp Hematol 2018; 63:52-63.e5. [PMID: 29656114 PMCID: PMC6371922 DOI: 10.1016/j.exphem.2018.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 11/15/2022]
Abstract
Dual insulin-like growth factor 1 receptor (IGF1/R) + mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibition synergistically sensitize apoptosis-resistant acute lymphoblastic leukemia (ALL) cells. Dual IGF1/R + MEK inhibition potentiates chemotherapy-induced killing of ALL cells. Signal transducer and activator of transcription 6 (STAT6) and platelet-derived growth factor-associated protein 1 (PDAP1) are downregulated differentially by this synergistic action. STAT6 and PDAP1 are predicted to act in a putative STAT6–ERK–nuclear factor kappa beta (NF-κB) survival network. Targeting this network may be useful for sensitizing ALL to chemotherapy agents.
Heterogeneous upregulation of multiple prosurvival pathways underlies resistance to damage-induced apoptosis in acute lymphoblastic leukemia (ALL) cells despite normal p53 responses. Here, we show that the dual combination of insulin-like growth factor 1 (IGF1)/IGF1 receptor (IGF1/R) and mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibition using AG1024 + U0126 can sensitize apoptosis-resistant ALL cells to ionizing radiation-induced DNA damage irrespective of effect of single pathway inhibition in vitro. This AG1024 + U0126 combination also significantly potentiates the ability of the core chemotherapy compounds vincristine, dexamethasone, and daunorubicin to kill ALL cells in vitro. Evidence of the synergistic action of AG1024 + U0126 in samples with variable basal levels of phosphorylated IGF1/Rβ and ERK1/2 suggested additional targets of this drug combination. Consistent with this, gene expression profiling identified 32 “synergy genes” differentially targeted by IGF1/R + MEK inhibition and, among these, Signal transducer and activator of transcription 6 (STAT6) and platelet-derived growth factor-associated protein 1 (PDAP1) were the most differentially downregulated cluster. Pearson correlation analysesrevealed that STAT6 and PDAP1 display significant expression codependency and a common expression pattern linked with other key “synergy” genes, supporting their predicted role in an STAT6–ERK–nuclear factor kappa beta (NF-κB) network. Knockdown studies revealed that loss of STAT6, but not PDAP1, impinges on the cell cycle, causing reduced numbers of viable cells. In combination with daunorubicin, STAT6 loss has an additive effect on cell killing, whereas PDAP1 loss is synergistic, indicating an important role of PDAP1 in the cellular response to this anthracycline. Inhibition of STAT6 or PDAP1 may therefore represent a potential novel therapeutic strategy for resistant ALL by enhancing sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Victoria J Weston
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Wenbin Wei
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Tatjana Stankovic
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Pamela Kearns
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
24
|
Fernando TR, Contreras JR, Zampini M, Rodriguez-Malave NI, Alberti MO, Anguiano J, Tran TM, Palanichamy JK, Gajeton J, Ung NM, Aros CJ, Waters EV, Casero D, Basso G, Pigazzi M, Rao DS. The lncRNA CASC15 regulates SOX4 expression in RUNX1-rearranged acute leukemia. Mol Cancer 2017; 16:126. [PMID: 28724437 PMCID: PMC5517805 DOI: 10.1186/s12943-017-0692-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/03/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play a variety of cellular roles, including regulation of transcription and translation, leading to alterations in gene expression. Some lncRNAs modulate the expression of chromosomally adjacent genes. Here, we assess the roles of the lncRNA CASC15 in regulation of a chromosomally nearby gene, SOX4, and its function in RUNX1/AML translocated leukemia. RESULTS CASC15 is a conserved lncRNA that was upregulated in pediatric B-acute lymphoblastic leukemia (B-ALL) with t (12; 21) as well as pediatric acute myeloid leukemia (AML) with t (8; 21), both of which are associated with relatively better prognosis. Enforced expression of CASC15 led to a myeloid bias in development, and overall, decreased engraftment and colony formation. At the cellular level, CASC15 regulated cellular survival, proliferation, and the expression of its chromosomally adjacent gene, SOX4. Differentially regulated genes following CASC15 knockdown were enriched for predicted transcriptional targets of the Yin and Yang-1 (YY1) transcription factor. Interestingly, we found that CASC15 enhances YY1-mediated regulation of the SOX4 promoter. CONCLUSIONS Our findings represent the first characterization of this CASC15 in RUNX1-translocated leukemia, and point towards a mechanistic basis for its action.
Collapse
Affiliation(s)
- Thilini R Fernando
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Jorge R Contreras
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Cellular and Molecular Pathology Ph.D. Program, UCLA, Los Angeles, USA
| | - Matteo Zampini
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Norma I Rodriguez-Malave
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Cellular and Molecular Pathology Ph.D. Program, UCLA, Los Angeles, USA.,Present Address: Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Michael O Alberti
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jaime Anguiano
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: University of San Francisco, 2130 Fulton St, San Francisco, CA, 94117, USA
| | - Tiffany M Tran
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Molecular, Cellular and Integrative Physiology Ph.D. program, UCLA, Los Angeles, USA
| | - Jayanth K Palanichamy
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Jasmine Gajeton
- Microbiology, Immunology and Molecular Genetics Program, UCLA, Los Angeles, USA.,Present Address: Department of Molecular Cardiology Lerner Research Institute, 9500 Euclid Avenue. Cleveland, Cleveland, OH, 44195, USA
| | - Nolan M Ung
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | - Cody J Aros
- Medical Scientist Training Program, David Geffen School of Medicine, UCLA, Los Angeles, USA
| | - Ella V Waters
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.,Present Address: Department of Molecular and Cell Biology, UC Berkeley, Berkeley, USA
| | - David Casero
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | - Giuseppe Basso
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Martina Pigazzi
- Women and Child Health Department- Hematology-Oncology laboratory, University of Padova, Padova, Italy
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA. .,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, USA. .,Broad Stem Cell Research Center, UCLA, 650 Charles E. Young Drive, Factor 12-272, Los Angeles, CA, 90095, USA.
| |
Collapse
|
25
|
Terwilliger T, Abdul-Hay M. Acute lymphoblastic leukemia: a comprehensive review and 2017 update. Blood Cancer J 2017; 7:e577. [PMID: 28665419 PMCID: PMC5520400 DOI: 10.1038/bcj.2017.53] [Citation(s) in RCA: 749] [Impact Index Per Article: 93.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/21/2017] [Indexed: 01/06/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the second most common acute leukemia in adults, with an incidence of over 6500 cases per year in the United States alone. The hallmark of ALL is chromosomal abnormalities and genetic alterations involved in differentiation and proliferation of lymphoid precursor cells. In adults, 75% of cases develop from precursors of the B-cell lineage, with the remainder of cases consisting of malignant T-cell precursors. Traditionally, risk stratification has been based on clinical factors such age, white blood cell count and response to chemotherapy; however, the identification of recurrent genetic alterations has helped refine individual prognosis and guide management. Despite advances in management, the backbone of therapy remains multi-agent chemotherapy with vincristine, corticosteroids and an anthracycline with allogeneic stem cell transplantation for eligible candidates. Elderly patients are often unable to tolerate such regimens and carry a particularly poor prognosis. Here, we review the major recent advances in the treatment of ALL.
Collapse
Affiliation(s)
- T Terwilliger
- New York University School of Medicine, New York, USA
| | - M Abdul-Hay
- New York University School of Medicine, New York, USA
- Department of Hematology, New York University Perlmutter Cancer Center, New York, USA
| |
Collapse
|
26
|
Athale UH, Gibson PJ, Bradley NM, Malkin DM, Hitzler J. Minimal Residual Disease and Childhood Leukemia: Standard of Care Recommendations From the Pediatric Oncology Group of Ontario MRD Working Group. Pediatr Blood Cancer 2016; 63:973-82. [PMID: 26914030 DOI: 10.1002/pbc.25939] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 01/09/2016] [Accepted: 01/12/2016] [Indexed: 01/03/2023]
Abstract
Minimal residual disease (MRD) is an independent predictor of relapse risk in children with leukemia and is widely used for risk-adapted treatment. This article summarizes current evidence supporting the use of MRD, including clinical significance, current international clinical practice, impact statement, and recommended indications. The proposed MRD recommendations have been endorsed by the MRD Working Group of the Pediatric Oncology Group of Ontario and provide the foundation for a strategy that aims at equitable access to MRD evaluation for children with leukemia.
Collapse
Affiliation(s)
- Uma H Athale
- Division of Hematology/Oncology, Hamilton Health Sciences, McMaster Children's Hospital, Hamilton, Ontario, Canada.,Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Paul J Gibson
- Division of Hematology/Oncology, Children's Hospital, London Health Sciences Centre, London, Ontario, Canada.,Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
| | - Nicole M Bradley
- Pediatric Oncology Group of Ontario (POGO), Toronto, Ontario, Canada
| | - David M Malkin
- Pediatric Oncology Group of Ontario (POGO), Toronto, Ontario, Canada.,Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Johann Hitzler
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
27
|
Ikoma MRV, Beltrame MP, Ferreira SIACP, Souto EX, Malvezzi M, Yamamoto M. Proposal for the standardization of flow cytometry protocols to detect minimal residual disease in acute lymphoblastic leukemia. Rev Bras Hematol Hemoter 2015; 37:406-13. [PMID: 26670404 PMCID: PMC4678914 DOI: 10.1016/j.bjhh.2015.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 07/23/2015] [Accepted: 07/27/2015] [Indexed: 01/26/2023] Open
Abstract
Minimal residual disease is the most powerful predictor of outcome in acute leukemia and is useful in therapeutic stratification for acute lymphoblastic leukemia protocols. Nowadays, the most reliable methods for studying minimal residual disease in acute lymphoblastic leukemia are multiparametric flow cytometry and polymerase chain reaction. Both provide similar results at a minimal residual disease level of 0.01% of normal cells, that is, detection of one leukemic cell in up to 10,000 normal nucleated cells. Currently, therapeutic protocols establish the minimal residual disease threshold value at the most informative time points according to the appropriate methodology employed. The expertise of the laboratory in a cancer center or a cooperative group could be the most important factor in determining which method should be used. In Brazil, multiparametric flow cytometry laboratories are available in most leukemia treatment centers, but multiparametric flow cytometry processes must be standardized for minimal residual disease investigations in order to offer reliable and reproducible results that ensure quality in the clinical application of the method. The Minimal Residual Disease Working Group of the Brazilian Society of Bone Marrow Transplantation (SBTMO) was created with that aim. This paper presents recommendations for the detection of minimal residual disease in acute lymphoblastic leukemia based on the literature and expertise of the laboratories who participated in this consensus, including pre-analytical and analytical methods. This paper also recommends that both multiparametric flow cytometry and polymerase chain reaction are complementary methods, and so more laboratories with expertise in immunoglobulin/T cell receptor (Ig/TCR) gene assays are necessary in Brazil.
Collapse
Affiliation(s)
| | | | | | | | | | - Mihoko Yamamoto
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | |
Collapse
|
28
|
Abstract
Childhood cancers are rare but an important cause of morbidity and mortality in children younger than 15 y of age. Common childhood malignancies include leukemias (commonest, 30-40%), brain tumors (20%) and lymphoma (12%) followed by neuroblastoma, retinoblastoma and tumors arising from soft tissues, bones and gonads. Leukemias, the commonest childhood cancer, arise from clonal proliferation of abnormal hematopoietic cells leading to disruption of normal marrow function and marrow failure. The various clinical manifestations of leukemia result from unregulated proliferation of the malignant clone and bone marrow failure. There are two main subtypes, the commoner, acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). A small proportion may have chronic myeloid leukemia (CML) and juvenile myelomonocytic leukemia (JMML). A systematic approach is necessary for diagnosis. Treatment should be initiated as early as possible to avoid complications. A timely referral to a cancer center must be done if facilities for diagnosis/treatment, management of complications and provision for supportive care are not available at the treating center.
Collapse
|
29
|
Abstract
Childhood cancers are rare but an important cause of morbidity and mortality in children younger than 15 y of age. Common childhood malignancies include leukemias (commonest, 30-40%), brain tumors (20%) and lymphoma (12%) followed by neuroblastoma, retinoblastoma and tumors arising from soft tissues, bones and gonads. Leukemias, the commonest childhood cancer, arise from clonal proliferation of abnormal hematopoietic cells leading to disruption of normal marrow function and marrow failure. The various clinical manifestations of leukemia result from unregulated proliferation of the malignant clone and bone marrow failure. There are two main subtypes, the commoner, acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). A small proportion may have chronic myeloid leukemia (CML) and juvenile myelomonocytic leukemia (JMML). A systematic approach is necessary for diagnosis. Treatment should be initiated as early as possible to avoid complications. A timely referral to a cancer center must be done if facilities for diagnosis/treatment, management of complications and provision for supportive care are not available at the treating center.
Collapse
Affiliation(s)
- Rachna Seth
- Division of Oncology, Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India,
| | | |
Collapse
|
30
|
Qiao SK, Guo XN, Ren JH, Zhang JN, Wang Y. Quantitative detection of the human cervical cancer oncogene for monitoring the minimal residual disease in acute leukemia. Exp Biol Med (Maywood) 2014; 240:128-34. [PMID: 25034723 DOI: 10.1177/1535370214543067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The human cervical cancer oncogene (HCCR) has been shown to be over-expressed in some solid tumors, and its function is involved in negative regulation of p53 tumor suppressor gene. However, the roles of HCCR in leukemia remain unclear. The present study is to investigate whether the expression levels of HCCR mRNA are associated with clinical prognosis in patients with acute leukemia (AL) and to explore the potential use as a biomarker for monitoring minimal residual disease (MRD) in AL. The mRNA levels of HCCR1 and HCCR2 were quantified by real-time reverse transcription polymerase chain reaction in bone marrow samples from 80 adult de novo AL patients and 20 normal healthy donors. The expressions of HCCR1 and HCCR2 were significantly higher in patients with acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) than those in healthy donors (P < 0.01), but there was no significant difference between AML and ALL (P > 0.05). Besides white blood cell count, we did not find any significant correlation between HCCR expression and clinical characteristics, such as age, sex, CD34 antigen expression, and response to chemotherapy. HCCR was monitored in 12 cases during remission and/or relapse. Significant reductions of both HCCR1 and HCCR2 mRNA levels were observed in patients who had achieved complete remission after chemotherapy but not in patients with non-responsive. However, an increased HCCR expression was detected in these patients who relapsed. Our findings suggest that HCCR gene is over-expressed in AL patients and may be as a useful biomarker for monitoring MRD in AL.
Collapse
Affiliation(s)
- Shu-Kai Qiao
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Xiao-Nan Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Jin-Hai Ren
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Jing-Nan Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Ying Wang
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
31
|
Garnache Ottou F, Chandesris MO, Lhermitte L, Callens C, Beldjord K, Garrido M, Bedin AS, Brouzes C, Villemant S, Rubio MT, Belanger C, Suarez F, Deau B, Lefrère F, Hermine O, Asnafi V, Varet B, Macintyre E. Peripheral blood 8 colour flow cytometry monitoring of hairy cell leukaemia allows detection of high-risk patients. Br J Haematol 2014; 166:50-9. [PMID: 24661013 DOI: 10.1111/bjh.12839] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/23/2014] [Indexed: 11/26/2022]
Abstract
Although purine analogues have significantly improved the outcome of hairy cell leukaemia (HCL) patients, 30-40% relapse, illustrating the need for minimal residual disease (MRD) markers that can aid personalized therapeutic management. Diagnostic samples from 34 HCL patients were used to design an 8-colour flow cytometry (8-FC) tube for blood MRD (B/RD) analysis (188 samples) which was compared to quantitative IGH polymerase chain reaction (Q-PCR) on 83 samples and to qualitative consensus IGH PCR clonality analysis on 165 samples. Despite heterogeneous HCL phenotypes at diagnosis, discrimination from normal B lymphocytes was possible in all cases using a single 8-FC tube, with a robust sensitivity of detection of 10(-4) , comparable to Q-PCR at this level, but preferable in terms of informativeness, simplicity and cost. B/RD assessment of 15 patients achieving haematological complete remission after purine analogues was predictive of a clinically significant relapse risk: with a median follow-up of 95 months; only one of the nine patients with reproducible 8-FC B/RD levels below 10(-4) (B/RD(neg) ) relapsed, compared to 5/6 in the B/RD(pos) group (P = 0.003). These data demonstrate the clinical interest of a robust 8-FC HCL B/RD strategy that could become a surrogate biomarker for therapeutic stratification and new drug assessment, which should be evaluated prospectively.
Collapse
Affiliation(s)
- Francine Garnache Ottou
- Laboratoire d'Hématologie and CNRS UMR8147, Hôpital Necker - Enfants Malades, Université Paris Descartes, 149 rue de Sèvres, 75743, Paris Cedex 15, France; INSERM UMR1098, Université de Franche-Comté, EFS-B/FC Plateforme de BioMonitoring, 1 Bd Fleming, 25000, Besançon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Tong H, Wang H, Wang Q, Liu Z, Lu C. Immunophenotypic, Cytogenetic and Clinical Features in Chinese Adult Acute Lymphoblastic Leukaemia (ALL) Patients. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2014. [DOI: 10.47102/annals-acadmedsg.v43n3p152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Introduction: This study sought to investigate the immunophenotypic subtype profiles of 110 Chinese adult patients with acute lymphoblastic leukaemia (ALL) and its association to cytogenetics and the clinical features. Materials and Methods: A total of 110 adult patients with ALL were immunophenotyped by CD45/SSC double parameters and 4 colour flow cytometry. Seventy-three cases were also subjected to karyotype analysis by R-banding technology. The clinical and laboratory data of 110 ALL patients were retrospectively analysed. Results: Of all the patients, 21.8% were identified as T-ALL, 78.2% as B-ALL. Abnormal karyotypes were detected in 37 out of 73 (50.7%) cases and the most common cytogenetic abnormality was the Philadelphia (Ph) chromosome, which was found in 23.3% (17/73) of the cases. Myeloid antigen (MyAg) expression was documented in 47.3% of the 110 adult ALL cases analysed and CD13 was the most commonly expressed MyAg in ALL patients (32.1 %). No difference was observed in the expression of MyAg between the groups of patients with T-ALL (45.8%) and B-ALL (47.7%). Our data showed that older age, higher CD34 positivity and lower proportion of patients with splenomegaly were found to be correlated with MyAg+ ALL, and that patients with Ph+ B-ALL were older, presented with higher haemoglobin level and higher CD34 expression. No statistical difference was noted in complete remission (CR) rate, relapse rate, induction mortality or total death rate among My+ and My-, Ph+ and Ph-, or B-ALL and T-ALL patients. Conclusion: Our results indicate that the distribution of ALL in Chinese adult patients was similar with the general distribution pattern in the other countries, and the expression of MyAg in patients with T-ALL and B-ALL was comparable. Both the expression of MyAg and the presence of Ph chromosome in adult ALL were significantly associated with median age and CD34 expression while not with the response to induction treatment.
Key words: Biological characteristics, Cytogenetics, Immunophenotype
Collapse
Affiliation(s)
- Haixia Tong
- Shengjing Hospital of China Medical University, People’s Republic of China
| | - Huihan Wang
- Shengjing Hospital of China Medical University, People’s Republic of China
| | - Qiushi Wang
- Shengjing Hospital of China Medical University, People’s Republic of China
| | - Zhuogang Liu
- Shengjing Hospital of China Medical University, People’s Republic of China
| | - Chunwei Lu
- College of Public Health, China Medical University, People’s Republic of China
| |
Collapse
|
33
|
Pati HP, Jain S. Flow cytometry in hematological disorders. Indian J Pediatr 2013; 80:772-8. [PMID: 23943573 DOI: 10.1007/s12098-013-1152-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 06/19/2013] [Indexed: 01/03/2023]
Abstract
Flow cytometry with its rapidly increasing applications is being used essentially in all fields of diagnostic medicine. In hematological disorders it is most commonly used in diagnosis, characterization, prognostication and even selecting target therapy of acute leukemia and to some extent lymphomas. It is increasingly finding place in other fields of hematology i.e., non-malignant disorders of all blood cell types including RBCs and platelets along with leukocytes. In this review the authors have discussed some of these applications with an emphasis on disorders specific to pediatric patients.
Collapse
Affiliation(s)
- Hara Prasad Pati
- Department of Hematology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| | | |
Collapse
|
34
|
BET inhibition as a single or combined therapeutic approach in primary paediatric B-precursor acute lymphoblastic leukaemia. Blood Cancer J 2013; 3:e126. [PMID: 23872705 PMCID: PMC3730202 DOI: 10.1038/bcj.2013.24] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/13/2013] [Accepted: 06/14/2013] [Indexed: 01/21/2023] Open
Abstract
Paediatric B-precursor ALL is a highly curable disease, however, treatment resistance in some patients and the long-term toxic effects of current therapies pose the need for more targeted therapeutic approaches. We addressed the cytotoxic effect of JQ1, a highly selective inhibitor against the transcriptional regulators, bromodomain and extra-terminal (BET) family of proteins, in paediatric ALL. We showed a potent in vitro cytotoxic response of a panel of primary ALL to JQ1, independent of their prognostic features but dependent on high MYC expression and coupled with transcriptional downregulation of multiple pro-survival pathways. In agreement with earlier studies, JQ1 induced cell cycle arrest. Here we show that BET inhibition also reduced c-Myc protein stability and suppressed progression of DNA replication forks in ALL cells. Consistent with c-Myc depletion and downregulation of pro-survival pathways JQ1 sensitised primary ALL samples to the classic ALL therapeutic agent dexamethasone. Finally, we demonstrated that JQ1 reduces ALL growth in ALL xenograft models, both as a single agent and in combination with dexamethasone. We conclude that targeting BET proteins should be considered as a new therapeutic strategy for the treatment of paediatric ALL and particularly those cases that exhibit suboptimal responses to standard treatment.
Collapse
|
35
|
Heel K, Tabone T, Röhrig KJ, Maslen PG, Meehan K, Grimwade LF, Erber WN. Developments in the immunophenotypic analysis of haematological malignancies. Blood Rev 2013; 27:193-207. [PMID: 23845589 DOI: 10.1016/j.blre.2013.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunophenotyping is the method by which antibodies are used to detect cellular antigens in clinical samples. Although the major role is in the diagnosis and classification of haematological malignancies, applications have expanded over the past decade. Immunophenotyping is now used extensively for disease staging and monitoring, to detect surrogate markers of genetic aberrations, to identify potential immuno-therapeutic targets and to aid prognostic prediction. This expansion in applications has resulted from developments in antibodies, methodology, automation and data handling. In this review we describe recent advances in both the technology and applications for the analysis of haematological malignancies. We highlight the importance of the expanding repertoire of testing capability for diagnostic, prognostic and therapeutic applications. The impact and significance of immunophenotyping in the assessment of haematological neoplasms are evident.
Collapse
Affiliation(s)
- Kathy Heel
- Pathology and Laboratory Medicine, University of Western Australia, Crawley, WA 6009, Australia.
| | | | | | | | | | | | | |
Collapse
|
36
|
Genome-wide association study identifies germline polymorphisms associated with relapse of childhood acute lymphoblastic leukemia. Blood 2012; 120:4197-204. [PMID: 23007406 DOI: 10.1182/blood-2012-07-440107] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
With the use of risk-directed therapy for childhood acute lymphoblastic leukemia (ALL), outcome has improved dramatically in the past 40 years. However, a substantial portion of patients, many of whom have no known risk factors, experience relapse. Taking a genome-wide approach, in the present study, we evaluated the relationships between genotypes at 444 044 single nucleotide polymorphisms (SNPs) with the risk of relapse in 2535 children with newly diagnosed ALL after adjusting for genetic ancestry and treatment regimen. We identified 134 SNPs that were reproducibly associated with ALL relapse. Of 134 relapse SNPs, 133 remained prognostic after adjusting for all known relapse risk factors, including minimal residual disease, and 111 were significant even among patients who were negative for minimal residual disease after remission induction therapy. The C allele at rs7142143 in the PYGL gene was associated with 3.6-fold higher risk of relapse than the T allele (P = 6.7 × 10(-9)). Fourteen of the 134 relapse SNPs, including variants in PDE4B and ABCB1, were also associated with antileukemic drug pharmacokinetics and/or pharmacodynamics. In the present study, we systematically identified host genetic variations related to treatment outcome of childhood ALL, most of which were prognostic independent of known risk factors for relapse, and some of which also influenced outcome by affecting host dis-position of antileukemic drugs. All trials are registered at www.clinicaltrials.gov or www.cancer.gov (COG P9904: NCT00005585; COG P9905: NCT00005596; COG P9906: NCT00005603; St Jude Total XIIIB: NCI-T93-0101D; and St Jude Total XV: NCT00137111).
Collapse
|
37
|
Abstract
The prognostic significance of minimal residual disease (MRD) has been demonstrated for a variety of hematologic malignancies. PCR based assays are among the most important methods for identifying MRD. They are aimed at detecting genetic abnormalities of residual leukemic cells with high specificity and sensitivity and represent an important diagnostic tool to assess the quality of therapeutic response, for clinical risk assessment, and for clinical management. In the present review technical aspects of different MRD detection methods are discussed which depend on the available targets regularly present in the respective leukemia type and subtype. As such fusion transcripts, gene mutations, and clonal rearrangements of antigen-receptor genes may be available for detection. Emphasis is given on discussing benefits and limitations of MRD detection and quantification in CML, AML and ALL.
Collapse
Affiliation(s)
- Javier Garcés-Eisele
- Molecular Biology Department, Laboratorios Clínicos de Puebla, and School of Health Sciences, Universidad Popular Autónoma del Estado de Puebla, México.
| |
Collapse
|
38
|
Liu X, Hsieh HB, Campana D, Bruce RH. A new method for high speed, sensitive detection of minimal residual disease. Cytometry A 2011; 81:169-75. [PMID: 21915990 DOI: 10.1002/cyto.a.21124] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 07/08/2011] [Accepted: 07/20/2011] [Indexed: 11/08/2022]
Abstract
Investigations of rare cell types in peripheral blood samples, such as tumor, fetal, and endothelial cells, represent an emerging field with several potentially valuable medical applications. Peripheral blood is a particularly attractive body fluid for the detection of rare cells as its collection is minimally invasive and can be repeated throughout the course of the disease. Because the number of rare cells in mononuclear cells can be very low (1 in 10 million), a large number of cells must be quickly screened, which places demanding requirements on the screening technology. While enrichment technology has shown promise in managing metastatic disease, enrichment can cause distortions of cell morphology that limit pathological identification, and the enrichment targeting adds additional constraints that can affect sensitivity. Here, we describe a new approach for detecting rare leukemia cells that does not require prior enrichment. We have developed an immunocytochemical assay for identification of leukemia cells spiked in peripheral blood samples, and a high-speed scanning instrument with high numerical aperture and wide field of view to efficiently locate these cells in large sample sizes. A multiplex immunoassay with four biomarkers was used to uniquely identify the rare cells from leukocytes and labeling artifacts. The cytometer preserves the cell morphology and accurately locates labeled rare cells for subsequent high resolution imaging. The sensitivity and specificity of the approach show promise for detection of a low number of leukemia cells in blood (1 in 10 million nucleated cells). The method enables rapid location of rare circulating cells (25 M cells/min), no specific enrichment step, and excellent imaging of cellular morphology with multiple immunofluorescent markers. The cell imaging is comparable to other imaging approaches such as laser scan cytometry and image flow cytometry, but the cell analysis rate is many orders of magnitude faster making this approach practical for detection of rare cells.
Collapse
Affiliation(s)
- Xiaohe Liu
- Biomedical Engineering, Palo Alto Research Center, Palo Alto, California, USA
| | | | | | | |
Collapse
|
39
|
Jiao Y, Cui L, Gao C, Li W, Zhao X, Liu S, Wu M, Deng G, Li Z. CASP8AP2 is a promising prognostic indicator in pediatric acute lymphoblastic leukemia. Leuk Res 2011; 36:67-71. [PMID: 21696825 DOI: 10.1016/j.leukres.2011.05.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 05/13/2011] [Accepted: 05/17/2011] [Indexed: 12/16/2022]
Abstract
The prognostic significance of caspase 8 associated protein 2 (CASP8AP2) in pediatric ALL is controversial. We determined a cut-off of CASP8AP2 expression in bone marrow samples of 39 newly diagnosed patients, and found a significantly poor bone marrow relapse-free survival (p=0.019) in low-expression group and verified it in another cohort of 106 patients (p=0.002). Furthermore, as an independent prognostic factor, CASP8AP2 expression was correlated to minimal residual disease (MRD), and incorporating it with MRD would help to identify patients at greater risk of bone marrow relapse. We also developed an algorithm comprised of clinical risk and CASP8AP2 expression, which could predict bone marrow relapse more accurately.
Collapse
Affiliation(s)
- Ying Jiao
- Key Laboratory of Major Diseases in Children, Ministry of Education & National Key Discipline of Pediatrics, Ministry of Education & Department of Hematology, Beijing Children's Hospital, Capital Medical University, 56 Nanlishi Road, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Topp MS, Kufer P, Gökbuget N, Goebeler M, Klinger M, Neumann S, Horst HA, Raff T, Viardot A, Schmid M, Stelljes M, Schaich M, Degenhard E, Köhne-Volland R, Brüggemann M, Ottmann O, Pfeifer H, Burmeister T, Nagorsen D, Schmidt M, Lutterbuese R, Reinhardt C, Baeuerle PA, Kneba M, Einsele H, Riethmüller G, Hoelzer D, Zugmaier G, Bargou RC. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol 2011; 29:2493-8. [PMID: 21576633 DOI: 10.1200/jco.2010.32.7270] [Citation(s) in RCA: 693] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Blinatumomab, a bispecific single-chain antibody targeting the CD19 antigen, is a member of a novel class of antibodies that redirect T cells for selective lysis of tumor cells. In acute lymphoblastic leukemia (ALL), persistence or relapse of minimal residual disease (MRD) after chemotherapy indicates resistance to chemotherapy and results in hematologic relapse. A phase II clinical study was conducted to determine the efficacy of blinatumomab in MRD-positive B-lineage ALL. PATIENTS AND METHODS Patients with MRD persistence or relapse after induction and consolidation therapy were included. MRD was assessed by quantitative reverse transcriptase polymerase chain reaction for either rearrangements of immunoglobulin or T-cell receptor genes, or specific genetic aberrations. Blinatumomab was administered as a 4-week continuous intravenous infusion at a dose of 15 μg/m2/24 hours. RESULTS Twenty-one patients were treated, of whom 16 patients became MRD negative. One patient was not evaluable due to a grade 3 adverse event leading to treatment discontinuation. Among the 16 responders, 12 patients had been molecularly refractory to previous chemotherapy. Probability for relapse-free survival is 78% at a median follow-up of 405 days. The most frequent grade 3 and 4 adverse event was lymphopenia, which was completely reversible like most other adverse events. CONCLUSION Blinatumomab is an efficacious and well-tolerated treatment in patients with MRD-positive B-lineage ALL after intensive chemotherapy. T cells engaged by blinatumomab seem capable of eradicating chemotherapy-resistant tumor cells that otherwise cause clinical relapse.
Collapse
Affiliation(s)
- Max S Topp
- Comprehensive Cancer Center Mainfranken, University Wuerzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
In vitro cellular drug resistance adds prognostic information to other known risk-factors in childhood acute lymphoblastic leukemia. Leuk Res 2011; 35:472-8. [DOI: 10.1016/j.leukres.2010.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 09/02/2010] [Accepted: 09/02/2010] [Indexed: 11/16/2022]
|
42
|
Margolin JF. Molecular diagnosis and risk-adjusted therapy in pediatric hematologic malignancies: a primer for pediatricians. Eur J Pediatr 2011; 170:419-25. [PMID: 21350806 DOI: 10.1007/s00431-011-1424-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Accepted: 02/08/2011] [Indexed: 12/17/2022]
Abstract
UNLABELLED Progress in the care of the hematologic malignancies of childhoond has been one of the proudest success stories in modern pediatrics. The cure rates of these diseases have improved from essentially zero in the 1950's and early 1960's to cure rates that range from 65%-90% in modern centers. While the largest improvements have been made in the most common (and the lower risk subtypes) of Acute Lymphoblastic Leukemia (ALL), there has also been significant progress in both the higher risk forms of ALL (i.e. Philadelphia chromosome positive, Ph+ ALL) and in Acute Myeloid Leukemia (AML). This progress has been achieved by the careful and stepwise identification of clinical, cytogenetic, molecular, and most recently response-based prognostic criteria, that now allow oncologists to focus the intensity of the therapy more closely to what is required to cure individual subgroups of patients. CONCLUSION Pediatricians need to be familiar with the changes in diagnostic and therapeutic approaches, because these changes have impact on: the laboratory tests that should be ordered at the time of specialist referral; counseling of patients and their families; and with the advent of "shared care models" pediatricians will need to be more involved in the general, supportive, and long-term care of these patients.
Collapse
|
43
|
Pui CH, Pei D, Campana D, Bowman WP, Sandlund JT, Kaste SC, Ribeiro RC, Rubnitz JE, Coustan-Smith E, Jeha S, Cheng C, Metzger ML, Bhojwani D, Inaba H, Raimondi SC, Onciu M, Howard SC, Leung W, Downing JR, Evans WE, Relling MV. Improved prognosis for older adolescents with acute lymphoblastic leukemia. J Clin Oncol 2011; 29:386-91. [PMID: 21172890 PMCID: PMC3058285 DOI: 10.1200/jco.2010.32.0325] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 09/15/2010] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The prognosis for older adolescents and young adults with acute lymphoblastic leukemia (ALL) has been historically much worse than that for younger patients. We reviewed the outcome of older adolescents (age 15 to 18 years) treated in four consecutive Total Therapy studies to determine if recent improved treatment extended to this high-risk group. PATIENTS AND METHODS Between 1991 and 2007, 963 pediatric patients, including 89 older adolescents, were enrolled on Total Therapy studies XIIIA, XIIIB, XIV, and XV. In the first three studies, treatment selection was based on presenting clinical features and leukemic cell genetics. In study XV, the level of residual disease was used to guide treatment, which featured intensive methotrexate, glucocorticoid, vincristine, and asparaginase, as well as early triple intrathecal therapy for higher-risk ALL. RESULTS The 89 older adolescents were significantly more likely to have T-cell ALL, the t(4;11)(MLL-AF4), and detectable minimal residual disease during or at the end of remission induction; they were less likely to have the t(12;21)(ETV6-RUNX1) compared with younger patients. In the first three studies, the 44 older adolescents had significantly poorer event-free survival and overall survival than the 403 younger patients. This gap in prognosis was abolished in study XV: event-free survival rates at 5 years were 86.4% ± 5.2% (standard error) for the 45 older adolescents and 87.4% ± 1.7% for the 453 younger patients; overall survival rates were 87.9% ± 5.1% versus 94.1% ± 1.2%, respectively. CONCLUSION Most older adolescents with ALL can be cured with risk-adjusted intensive chemotherapy without stem-cell transplantation.
Collapse
Affiliation(s)
- Ching-Hon Pui
- St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chetverina EV, Chetverin AB. Nanocolonies and diagnostics of oncological diseases associated with chromosomal translocations. BIOCHEMISTRY (MOSCOW) 2011; 75:1667-91. [DOI: 10.1134/s0006297910130109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Recent research advances in childhood acute lymphoblastic leukemia. J Formos Med Assoc 2011; 109:777-87. [PMID: 21126650 DOI: 10.1016/s0929-6646(10)60123-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 08/01/2010] [Accepted: 08/02/2010] [Indexed: 01/26/2023] Open
Abstract
Recent progress in risk-adapted treatment for childhood acute lymphoblastic leukemia has secured 5-year event-free survival rates of approximately 80% and 5-year survival rates approaching 90%. With improved systemic and intrathecal chemotherapy, it is now feasible to omit safely in all patients prophylactic cranial irradiation, which was once a standard treatment. As high-resolution, genome-wide analyses of leukemic and normal host cells continue to identify novel subtypes of lymphoblastic leukemia and provide new insights into leukemogenesis, we can look forward to the time when all cases of this disease will be classified according to specific genetic abnormalities, some of which will yield "druggable" targets for more effective and less toxic treatments. Meanwhile, it is sobering to consider that a significant fraction of leukemia survivors will develop serious health problems within 30 years of their initial diagnosis. This underlines the need to introduce early countermeasures to reduce late therapy-related effects. The ultimate challenge is to gain a clear understanding of the factors that give rise to childhood leukemia in the first place, and enable preventive strategies to be devised and implemented.
Collapse
|
46
|
Koh KN, Park M, Kim BE, Im HJ, Park CJ, Jang S, Chi HS, Seo JJ. Prognostic significance of minimal residual disease detected by a simplified flow cytometric assay during remission induction chemotherapy in children with acute lymphoblastic leukemia. KOREAN JOURNAL OF PEDIATRICS 2010; 53:957-64. [PMID: 21218018 PMCID: PMC3012276 DOI: 10.3345/kjp.2010.53.11.957] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 08/27/2010] [Accepted: 09/13/2010] [Indexed: 01/22/2023]
Abstract
Purpose Our study attempted to determine the prognostic significance of minimal residual disease (MRD) detected by a simplified flow cytometric assay during induction chemotherapy in children with B-cell acute lymphoblastic leukemia (B-ALL). Methods A total of 98 patients were newly diagnosed with precursor B-ALL from June 2004 to December 2008 at the Asan Medical Center (Seoul, Korea). Of those, 37 were eligible for flow cytometric MRD study analysis on day 14 of their induction treatment. The flow cytometric MRD assay was based on the expression intensity of CD19/CD10/CD34 or aberrant expression of myeloid antigens by bone marrow nucleated cells. Results Thirty-five patients (94.6%) had CD19-positive leukemic cells that also expressed CD10 and/or CD34, and 18 (48.6%) had leukemic cells with aberrant expression of myeloid antigens. Seven patients with ≥1% leukemic cells on day 14 had a significantly lower relapse-free survival (RFS) compared to the 30 patients with lower levels (42.9% [18.7%] vs. 92.0% [5.4%], P=0.004). Stratification into 3 MRD groups (≥1%, 0.1-1%, and <0.1%) also showed a statistically significant difference in RFS (42.9% [18.7%] vs. 86.9% [8.7%] vs. 100%, P=0.013). However, the MRD status had no significant influence on overall survival. Multivariate analysis demonstrated that the MRD level on day 14 was an independent prognostic factor with borderline significance. Conclusion An MRD assay using simplified flow cytometry during induction chemotherapy may help to identify patients with B-ALL who have an excellent outcome and patients who are at higher risk for relapse.
Collapse
Affiliation(s)
- Kyung Nam Koh
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Kikuchi M, Tanaka J, Kondo T, Hashino S, Kasai M, Kurosawa M, Iwasaki H, Morioka M, Kawamura T, Masauzi N, Fukuhara T, Kakinoki Y, Kobayashi H, Noto S, Asaka M, Imamura M. Clinical significance of minimal residual disease in adult acute lymphoblastic leukemia. Int J Hematol 2010; 92:481-9. [DOI: 10.1007/s12185-010-0670-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 07/23/2010] [Accepted: 08/17/2010] [Indexed: 10/19/2022]
|
48
|
Abstract
Treatment of acute lymphoblastic leukaemia (ALL) in adults presents a formidable challenge. While overall results have improved over the past 3 decades, the long-term survival for patients aged less than 60 years is only in the range of 30-40% and is 10-15% if between 60 and 70 years and <5% for those over 70 years. The historic lack of clear-cut biological prognostic factors has led to over- or under-treatment of some patients. Response to initial therapy is an important prognosticator of outcome based on disease biology, as well as pharmacogenetics, which include the patient's response to drugs given. The more widespread availability of allogeneic transplantation and reduced-intensity regimens for older patients have opened up this curative modality to a greater number of patients. Hopefully, those options, as well as novel cytogenetic and molecular markers, will enable a better selection of patients who undergo intensive therapies and finally break the 30-40% cure barrier for adults with ALL.
Collapse
Affiliation(s)
- Jacob M Rowe
- Rambam Health Care Campus and Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
49
|
Kröger N, Bacher U, Bader P, Böttcher S, Borowitz MJ, Dreger P, Khouri I, Macapinlac HA, Macapintac H, Olavarria E, Radich J, Stock W, Vose JM, Weisdorf D, Willasch A, Giralt S, Bishop MR, Wayne AS. NCI First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: report from the Committee on Disease-Specific Methods and Strategies for Monitoring Relapse following Allogeneic Stem Cell Transplantation. Part I: Methods, acute leukemias, and myelodysplastic syndromes. Biol Blood Marrow Transplant 2010; 16:1187-211. [PMID: 20558311 DOI: 10.1016/j.bbmt.2010.06.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 06/06/2010] [Indexed: 12/14/2022]
Abstract
Relapse has become the major cause of treatment failure after allogeneic stem cell transplantation. Outcome of patients with clinical relapse after transplantation generally remains poor, but intervention prior to florid relapse improves outcome for certain hematologic malignancies. To detect early relapse or minimal residual disease, sensitive methods such as molecular genetics, tumor-specific molecular primers, fluorescein in situ hybridization, and multiparameter flow cytometry (MFC) are commonly used after allogeneic stem cell transplantation to monitor patients, but not all of them are included in the commonly employed disease-specific response criteria. The highest sensitivity and specificity can be achieved by molecular monitoring of tumor- or patient-specific markers measured by polymerase chain reaction-based techniques, but not all diseases have such targets for monitoring. Similar high sensitivity can be achieved by determination of donor chimerism, but its specificity regarding detection of relapse is low and differs substantially among diseases. Here, we summarize the current knowledge about the utilization of such sensitive monitoring techniques based on tumor-specific markers and donor cell chimerism and how these methods might augment the standard definitions of posttransplant remission, persistence, progression, relapse, and the prediction of relapse. Critically important is the need for standardization of the different residual disease techniques and to assess the clinical relevance of minimal residual disease and chimerism surveillance in individual diseases, which in turn, must be followed by studies to assess the potential impact of specific interventional strategies.
Collapse
Affiliation(s)
- Nicolaus Kröger
- Department for Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinstrasse 52, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
LI XIN, DU WEN, LIU WEI, LI XIAOQING, LI HONGRUI, HUANG SHIANG. Comprehensive flow cytometry phenotype in acute leukemia at diagnosis and at relapse. APMIS 2010; 118:353-9. [DOI: 10.1111/j.1600-0463.2010.02603.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|