1
|
Yang H, Xun Y, Shen Y, Wang H, Tao Y, Wang H, Zhang X, Liu R, Yu H, Wei L, Yan J, Zhu X, You H. A simplified and robust risk stratification model for stem cell transplantation in pediatric acute myeloid leukemia. Cell Rep Med 2024; 5:101762. [PMID: 39366384 PMCID: PMC11513827 DOI: 10.1016/j.xcrm.2024.101762] [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: 12/13/2023] [Revised: 06/20/2024] [Accepted: 09/11/2024] [Indexed: 10/06/2024]
Abstract
The efficacy of stem cell transplantation (SCT) in pediatric acute myeloid leukemia (pAML) remains unsatisfactory due to the limitations of existing prognostic models in predicting efficacy and selecting suitable candidates. This study aims to develop a cytomolecular risk stratification-independent prognostic model for SCT in pAML patients at CR1 stage. The pAML SCT model, based on age, KMT2A rearrangement (KMT2A-r), and minimal residual disease at end of course 1 (MRD1), effectively classifies patients into low-, intermediate-, and high-risk groups. We validate the effectiveness in an internal validation cohort and in four external validation cohorts, consisting of different graft sources and donors. Moreover, by incorporating the FMS-like tyrosine kinase 3/internal tandem duplication (FLT3/ITD) allelic ratio, the pAML SCT model is refined, enhancing its ability to effectively select suitable candidates. We develop a simple and robust risk stratification model for pAML patients undergoing SCT, to aid in risk stratification and inform pretransplant decision-making at CR1 stage.
Collapse
Affiliation(s)
- Hua Yang
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Basic Medicine, School of Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Yang Xun
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Basic Medicine, School of Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Yali Shen
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongtao Wang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tao
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Huihan Wang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinyue Zhang
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Rongqiu Liu
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Huarong Yu
- College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Li Wei
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, China.
| | - Jinsong Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, China.
| | - Xiaoyu Zhu
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Hua You
- Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Department of Pediatric Hematology and Oncology, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Won DI, Lim JH, Cho JH, Kim CD, Yun WS, Huh S. 4-Color Flow Cytometric Crossmatch Using Whole Blood Lysis. Transplant Proc 2024; 56:1483-1492. [PMID: 38981762 DOI: 10.1016/j.transproceed.2024.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND In lymphocyte crossmatch using flow cytometry (flow cytometric crossmatch, FCXM), the conventional tricolor FCXM protocol requires a mononuclear cell isolation step. To develop a new, more streamlined protocol, we introduced whole blood lysis (WBL) and CD45 fluorescence-triggered acquisition using 4-color flow cytometry. METHODS A total of 186 donor/recipient pairs for transplantation were classified into donor-specific human leukocyte antigen (HLA) alloantibody-positive (DSA+, n = 78) and DSA-negative (DSA-, n = 108) groups. The latter group was reclassified into blood group ABO-incompatible (ABOi, n = 56) and ABO-compatible (n = 52) subgroups. The WBL FCXM protocol with CD45 V500-C was optimized using a FACSLyric cytometer (BD Biosciences) with 3 lasers. Measurements for T cells or B cells were calculated as a mean fluorescence intensity (MFI) ratio (test divided by control). WBL FCXM was compared with conventional FCXM in each group. RESULTS WBL FCXM showed no difference quantitatively compared with conventional FCXM, except for the B cell FCXM in the DSA- group (B cell MFI ratio: 1.06 ± 0.44 and 0.92 ± 0.41, respectively [P = .0001]). There was no ABO antibody interference in the ABOi subgroup. Similar results were observed in the qualitative determinations of FCXM as follows: 1) In the DSA+ group, the sensitivity of B cell WBL FCXM (96.2%) showed no difference compared with that of conventional FCXM (91.0%, P = .2188) and 2) In the DSA- group, the specificity of T cell WBL FCXM (96.3%) showed no difference compared with that of conventional FCXM (98.1%, P = .6250). WBL FCXM reduced the turnaround time by 50 min compared with that by conventional FCXM. CONCLUSIONS WBL FCXM demonstrated comparable assay performance to that of conventional FCXM. Because this new FCXM protocol is simple and does not compromise assay sensitivity, it has the potential to replace the conventional method in histocompatibility laboratory settings.
Collapse
Affiliation(s)
- Dong Il Won
- Department of Clinical Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| | - Jeong-Hoon Lim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jang-Hee Cho
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chan-Duck Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Woo-Sung Yun
- Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Huh
- Department of Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
3
|
Tatarian J, Tupper N, Li P, Feusier J, Abdo M, Hyter S, Gonzales PR, Zhang D, Woodroof J, Kelting S, Godwin AK, Cui W. Morphologic, immunophenotypic, molecular genetic, and clinical characterization in patients with SRSF2-mutated acute myeloid leukemia. Am J Clin Pathol 2023; 160:490-499. [PMID: 37458189 PMCID: PMC10629464 DOI: 10.1093/ajcp/aqad077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/01/2023] [Indexed: 09/21/2023] Open
Abstract
OBJECTIVES SRSF2 mutations are known to be associated with poor outcomes in myelodysplastic neoplasm, but studies on their prognostic impact on acute myeloid leukemia (AML) remain limited. In this retrospective study, we analyzed clinical and pathologic characteristics of patients with AML and correlated the outcomes with SRSF2 mutations. METHODS We characterized the morphologic, immunophenotypic, molecular, and clinical findings in AML with mutated SRSF2 and compared them with SRSF2 wild-type (WT) myeloid neoplasms (MNs). RESULTS Using next-generation sequencing, we identified 134 patients with MNs and SRSF2 mutations (85 with AML and 49 with MNs) in addition to 342 SRSF2-WT AMLs. Fifty-two (62%) patients with altered SRSF2 demonstrated a variable degree of morphologic dysplasia. The most frequent immunophenotypic aberrancies in SRSF2-mutant AML included diminished CD33 expression and overexpression of CD7, CD56, or CD123, similar to WT AML. More IDH1/2 (P = .015) and NPM1 (P = .002) mutations were seen in SRSF2-mutant AML than in SRSF2-mutant non-AML. Further, more IDH1/2, ASXL1, RUNX1, and STAG2 mutations were observed in SRSF2-mutant AML than in SRSF2-WT AML (P < .0001 to P = .001). Finally, patients with SRSF2-mutant AML showed a significantly worse overall survival (OS) than patients with SRSF2-WT AML (P < .0001), but this worse OS appeared to be rescued by allogeneic stem cell transplant (allo-SCT). CONCLUSIONS Acute myeloid leukemia with altered SRSF2 shows a variable degree of morphologic dysplasia without uniform immunophenotypic aberrancies. SRSF2 mutations appear to be independent poor prognostic factors, but allo-SCT has improved the clinical outcomes in patients with SRSF2-mutant AML.
Collapse
Affiliation(s)
- Joshua Tatarian
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Natalie Tupper
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Peng Li
- Division of Hematopathology, Department of Pathology, University of Utah, Salt Lake City, UT, US
| | - Julie Feusier
- Division of Hematopathology, Department of Pathology, University of Utah, Salt Lake City, UT, US
| | - Maryam Abdo
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Stephen Hyter
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Patrick R Gonzales
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Da Zhang
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Janet Woodroof
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Sarah Kelting
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Andrew K Godwin
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS, US
| | - Wei Cui
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, US
| |
Collapse
|
4
|
Li Y, Wang S, Xiao H, Lu F, Zhang B, Zhou T. Evaluation and validation of the prognostic value of platelet indices in patients with leukemia. Clin Exp Med 2023; 23:1835-1844. [PMID: 36622510 DOI: 10.1007/s10238-022-00985-z] [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: 12/10/2022] [Accepted: 12/29/2022] [Indexed: 01/10/2023]
Abstract
Platelets (PLTs) are believed to play a role in the process by which tumors can accelerate their growth rate, as well as offer the physical and mechanical support necessary to evade the immunological system and metastasis. There is, however, no literature available if PLTs have a role in leukemia. It is significant for PLTs to play a part in hematological malignancies from a therapeutic standpoint and to have the capacity to serve as a prognostic marker in the evolution of leukemia. This is because PLTs play a crucial role in the development of cancer and tumors. In this study, it will be shown that PLT count can be used to predict long-term prognosis after chemotherapy especially in the case of acute myeloid leukemia patients. Furthermore, low PLT-to-lymphocyte ratio and mean PLT volume, as well as high PLT distribution width, are associated with poor prognosis and may represent a novel independent prognostic factor.
Collapse
Affiliation(s)
- Yuyan Li
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Shuangge Wang
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Han Xiao
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Fang Lu
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Bin Zhang
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Tingting Zhou
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China.
| |
Collapse
|
5
|
Wang SA, Jorgensen JL, Hu S, Jia F, Li S, Loghavi S, Ok CY, Thakral B, Xu J, Medeiros LJ, Wang W. Validation of a 12-color flow cytometry assay for acute myeloid leukemia minimal/measurable residual disease detection. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:356-366. [PMID: 37605812 DOI: 10.1002/cyto.b.22140] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) minimal/measurable residual disease (MRD) by multicolor flow cytometry is a complex laboratory developed test (LDT), challenging for implementation. We share our experience in the validation of a 12-color AML MRD flow cytometry assay to meet stringent regulatory requirements. METHODS We worked under the guidelines of the CLSI HL62 publication, illustrated the details of the validation process that was tailored to uniqueness of AML MRD, and tested its clinical validity in 61 patients. The "trueness" was determined by correlating with concurrent molecular genetic testing and follow-up bone marrow examinations. RESULTS Under assay specificity, we shared the details of panel design, analysis, and criteria for interpretation and reporting. The assay accuracy was assessed by testing known positive and negative samples and correlating with molecular genetic testing and follow-up bone marrow examination. The limit of detection (LOD) and limit of quantification (LOQ) were validated to a level between 0.01% and 0.1%, varied from the leukemia-associated immunophenotypes (LAIP) and the numbers of events obtained for analysis. Assay linearity, precision and carry over studies all met acceptable criteria. In the clinical validity test, the concordance was 93%, specificity 98% and sensitivity 83%. The most challenging aspects of the assay were the discrimination of pre-leukemic cells (persistent clonal hematopoiesis) or underlying myelodysplastic clones from AML MRD with immunophenotypic switch or subclone selection. CONCLUSION The validation met all criteria and obtained FDA IDE (investigational device exemption) approval. This study provides ample technical and professional details in setting up the AML MRD flow cytometry assay and illustrates through the example of the "fit for purpose" validation process. We also highlight the need for further characterization of abnormal blasts bearing the potential for AML relapse.
Collapse
Affiliation(s)
- Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey L Jorgensen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shimin Hu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fuli Jia
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shaoying Li
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chi Young Ok
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Beenu Thakral
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jie Xu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
6
|
Wang Y, Quesada AE, Zuo Z, Medeiros LJ, Yin CC, Li S, Xu J, Borthakur G, Li Y, Yang C, Abaza Y, Gao J, Lu X, You MJ, Zhang Y, Lin P. The Impact of Mutation of Myelodysplasia-Related Genes in De Novo Acute Myeloid Leukemia Carrying NPM1 Mutation. Cancers (Basel) 2022; 15:cancers15010198. [PMID: 36612194 PMCID: PMC9818485 DOI: 10.3390/cancers15010198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Background: The impact of gene mutations typically associated with myelodysplastic syndrome (MDS) in acute myeloid leukemia (AML) with NPM1 mutation is unclear. Methods: Using a cohort of 107 patients with NPM1-mutated AML treated with risk-adapted therapy, we compared survival outcomes of patients without MDS-related gene mutations (group A) with those carrying concurrent FLT3-ITD (group B) or with MDS-related gene mutations (group C). Minimal measurable disease (MMD) status assessed by multiparameter flow cytometry (MFC), polymerase chain reaction (PCR), and/or next-generation sequencing (NGS) were reviewed. Results: Among the 69 patients treated intensively, group C showed significantly inferior progression-free survival (PFS, p < 0.0001) but not overall survival (OS, p = 0.055) compared to group A. Though groups A and C had a similar MMD rate, group C patients had a higher relapse rate (p = 0.016). Relapse correlated with MMD status at the end of cycle 2 induction (p = 0.023). Survival of group C patients was similar to that of group B. Conclusion: MDS-related gene mutations are associated with an inferior survival in NPM1-mutated AML.
Collapse
Affiliation(s)
- Yi Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andres E. Quesada
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhuang Zuo
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - L. Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - C. Cameron Yin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaoying Li
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Xu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yisheng Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chao Yang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasmin Abaza
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Juehua Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xinyan Lu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - M. James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yizhuo Zhang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Correspondence: (Y.Z.); (P.L.); Tel.: +86-18622221239 (Y.Z.); +1-(713)-794-1746 (P.L.); Fax: +86-022-23340123 (Y.Z.); +1-(713)-563-2977 (P.L.)
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (Y.Z.); (P.L.); Tel.: +86-18622221239 (Y.Z.); +1-(713)-794-1746 (P.L.); Fax: +86-022-23340123 (Y.Z.); +1-(713)-563-2977 (P.L.)
| |
Collapse
|
7
|
Gao L, Hyter S, Zhang D, Kelting S, Woodroof J, Abdallah AO, Yacoub A, McGuirk J, Abdelhakim H, Godwin AK, Cui W. Morphologic, immunophenotypic, and molecular genetic comparison study in patients with clonal cytopenia of undetermined significance, myelodysplastic syndrome, and acute myeloid leukemia with myelodysplasia-related changes: A single institution experience. Int J Lab Hematol 2022; 44:738-749. [PMID: 35352484 DOI: 10.1111/ijlh.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/15/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Next-generation sequencing (NGS) analysis showed clonal cytopenia of undetermined significance (CCUS) as an immediate precursor to myelodysplastic syndrome (MDS). METHODS We evaluated and compared morphologic, multiparametric flow cytometry (MFC), and molecular genetic findings in patients with CCUS (n = 37), MDS (n = 75), and acute myeloid leukemia with myelodysplasia-related changes (AML-MRC, n = 24). RESULTS CCUS patients showed variable MFC abnormalities including >2% CD34+ myeloblasts (5.8%), altered antigen expression on myeloblasts, monocytes, and granulocytes (1.2, 1.5, and 0.2/case), abnormal maturation of myeloblasts (45.8%), decreased hematogones (17.6%), and decreased side scatter (SSC) of granulocytes (11.4%). CCUS patients with high-risk mutations showed significantly more MFC abnormalities. However, CCUS patients with >20% variant allelic fraction (VAF) did not show more MFC aberrations than the rest of the group. MDS patients showed significantly more MFC abnormalities compared with CCUS patients (p = 7.8E-05-0.047). Low-grade MDS patients showed significantly fewer MFC abnormalities compared with high-grade MDS or AML-MRC patients (p = 1.89E-05-0.04). AML-MRC patients showed significantly elevated blast counts, more antigen aberrations, decreased hematogones, and decreased SSC of granulocytes compared with CCUS patients (p = 2.0E-05-0.01). CCUS patients carried predominantly TET2/DNMT3A/ASXL1 mutations. They harbored fewer mutations in gene coding splicing factors compared with MDS patients (p = .0001-.02) and fewer mutations in tumor suppressor and transcription factor genes compared with AML-MRC patients (p = .0006-.02). CONCLUSIONS CCUS is an immediate precursor to low-grade MDS. The progression from CCUS to MDS to AML-MRC is a stepwise process that requires acquisition of mutations in splicing, transcription factor, and tumor suppressor genes with accumulations of additional MFC abnormalities.
Collapse
Affiliation(s)
- Linlin Gao
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Stephen Hyter
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Da Zhang
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sarah Kelting
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Janet Woodroof
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Al-Ola Abdallah
- Division of Hematologic Malignancies & Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas, USA
| | - Abdulraheem Yacoub
- Division of Hematologic Malignancies & Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas, USA
| | - Joseph McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas, USA
| | - Haitham Abdelhakim
- Division of Hematologic Malignancies & Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Westwood, Kansas, USA
| | - Andrew K Godwin
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wei Cui
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
8
|
Dai P, Wu LR, Chen SX, Wang MX, Cheng LY, Zhang JX, Hao P, Yao W, Zarka J, Issa GC, Kwong L, Zhang DY. Calibration-free NGS quantitation of mutations below 0.01% VAF. Nat Commun 2021; 12:6123. [PMID: 34675197 PMCID: PMC8531361 DOI: 10.1038/s41467-021-26308-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/30/2021] [Indexed: 12/29/2022] Open
Abstract
Quantitation of rare somatic mutations is essential for basic research and translational clinical applications including minimal residual disease (MRD) detection. Though unique molecular identifier (UMI) has suppressed errors for rare mutation detection, the sequencing depth requirement is high. Here, we present Quantitative Blocker Displacement Amplification (QBDA) which integrates sequence-selective variant enrichment into UMI quantitation for accurate quantitation of mutations below 0.01% VAF at only 23,000X depth. Using a panel of 20 genes recurrently altered in acute myeloid leukemia, we demonstrate quantitation of various mutations including single base substitutions and indels down to 0.001% VAF at a single locus with less than 4 million sequencing reads, allowing sensitive MRD detection in patients during complete remission. In a pan-cancer panel and a melanoma hotspot panel, we detect mutations down to 0.1% VAF using only 1 million reads. QBDA provides a convenient and versatile method for sensitive mutation quantitation using low-depth sequencing.
Collapse
Affiliation(s)
- Peng Dai
- Department of Bioengineering, Rice University, Houston, TX, USA
- NuProbe USA, Houston, TX, USA
| | - Lucia Ruojia Wu
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Sherry Xi Chen
- Department of Bioengineering, Rice University, Houston, TX, USA
- NuProbe USA, Houston, TX, USA
| | | | | | | | | | | | - Jabra Zarka
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lawrence Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Yu Zhang
- Department of Bioengineering, Rice University, Houston, TX, USA.
- NuProbe USA, Houston, TX, USA.
| |
Collapse
|
9
|
F McGowan P, D Hyter S, Cui W, Plummer RM, Godwin AK, Zhang D. Comparison of flow cytometry and next-generation sequencing in minimal residual disease monitoring of acute myeloid leukemia: One institute's practical clinical experience. Int J Lab Hematol 2021; 44:118-126. [PMID: 34585519 DOI: 10.1111/ijlh.13711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/19/2021] [Accepted: 09/09/2021] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Monitoring patients with acute myeloid leukemia can be implemented through various techniques such as multiparameter flow cytometry, real-time quantitative polymerase chain reaction, and next-generation sequencing. However, there is scarce studies when comparing the data of next-generation sequencing and flow cytometry for monitoring disease progression, particularly how they might supplement one another when used in tandem. METHODS We investigated 107 patients via retrospective analysis using follow-up MFC and NGS data with a total of 717 MFC and 247 NGS studies to compare these methods in monitoring minimal/measurable residual disease. RESULTS 197 instances were MFC+ /NGS+ , 3 were MFC- /NGS- , 44 were MFC- /NGS+ , and 3 are MFC+ /NGS- . The majority of the MFC- /NGS+ cases occurred within 6 months during the post-treatment phase (64%). Among 44 MFC- /NGS+ instances, 13 had similar NGS profiles to their original day 0 diagnosis. The remaining cases showed preleukemic clonal hematopoiesis mutations, "likely pathogenic mutations," or "variants of uncertain significance." CONCLUSION Our findings show that flow cytometry has its advantages with comparable sensitivity in detecting minimal/measurable residual disease. Next-generation sequencing could be used in an increased and more regular capacity in conjunction with flow cytometry to achieve a more comprehensive surveillance of these patients, resulting in improved outcomes.
Collapse
Affiliation(s)
- Paul F McGowan
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Stephen D Hyter
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wei Cui
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Regina M Plummer
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Andrew K Godwin
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Da Zhang
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
10
|
Prognostic impact of complete remission with MRD negativity in patients with relapsed or refractory AML. Blood Adv 2021; 4:6117-6126. [PMID: 33351107 DOI: 10.1182/bloodadvances.2020002811] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
In relapsed/refractory acute myeloid leukemia (AML), the prognostic impact of complete remission (CR) and measurable residual disease (MRD) negativity is not well established. We retrospectively analyzed 141 patients with relapsed/refractory AML who received first salvage therapy and had MRD assessed by multiparameter flow cytometry at the time of response. Patients who achieved CR with full hematologic recovery as best response vs those with incomplete hematology recovery had lower cumulative incidence of relapse (P = .01) and better relapse-free survival (P = .004) but not overall survival (P = .15); a similar trend was observed in patients who achieved MRD negativity vs those who were MRD positive (P = .01, P = .05, and P = .21, respectively). By multivariate analysis, CR and MRD negativity were each independently associated with lower cumulative incidence of relapse (P = .001 and P = .003, respectively) and better relapse-free survival (P < .001 and P = .02) but not overall survival. Patients who achieved CR with MRD negativity had the lowest rates of relapse and best survival (2-year overall survival rate, 37%), which was driven largely by lower rates of early relapse and an increased ability in this group to undergo hematopoietic stem cell transplantation (HSCT); however, post-HSCT outcomes were similar regardless of response to salvage chemotherapy. Overall, in patients with relapsed/refractory AML, CR with MRD negativity was associated with the best outcomes, supporting it as the optimal response in this setting.
Collapse
|
11
|
Loghavi S, DiNardo CD, Furudate K, Takahashi K, Tanaka T, Short NJ, Kadia T, Konopleva M, Kanagal-Shamanna R, Farnoud NR, Pierce S, Khoury JD, Jorgensen JL, Patel KP, Daver N, Yilmaz M, Medeiros LJ, Kantarjian H, Ravandi F, Wang SA. Flow cytometric immunophenotypic alterations of persistent clonal haematopoiesis in remission bone marrows of patients with NPM1-mutated acute myeloid leukaemia. Br J Haematol 2021; 192:1054-1063. [PMID: 33618432 DOI: 10.1111/bjh.17347] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/22/2020] [Accepted: 12/14/2020] [Indexed: 01/07/2023]
Abstract
Clonal haematopoiesis (CH) in patients with acute myeloid leukaemia (AML) may persist beyond attaining complete remission. From a consecutive cohort of 67 patients with nucleophosmin 1-mutated (NPM1mut ) AML, we identified 50 who achieved NPM1mut clearance and had parallel multicolour flow cytometry (MFC) and next generation sequencing (NGS). In total, 13 (26%) cleared all mutations, 37 (74%) had persistent CH frequently involving DNA methyltransferase 3α (DNMT3A,70%), tet methylcytosine dioxygenase 2 (TET2, 27%), isocitrate dehydrogenase 2 (IDH2, 19%) and IDH1 (11%). A small number (<1%) of aberrant CD34+ myeloblasts, but immunophenotypically different from original AML blasts [herein referred to as a pre-leukaemic (PL) phenotype], was detected in 17 (49%) patients with CH, but not in any patients with complete clearance of all mutations (P = 0·0037). A PL phenotype was associated with higher mutation burden (P = 0·005). Persistent IDH2 and serine and arginine-rich splicing factor 2 (SRSF2) mutations were exclusively observed in PL+ CH+ cases (P = 0·016). Persistent dysplasia was seen exclusively in cases with a PL+ phenotype (29% vs. none; P = 0·04). The PL+ phenotype did not correlate with age, intensity of induction therapy or relapse-free survival. Post-remission CH in the setting of NPM1mut clearance is common and may result in immunophenotypic changes in myeloid progenitors. It is important to not misinterpret these cells as AML measurable residual disease (MRD).
Collapse
Affiliation(s)
- Sanam Loghavi
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Ken Furudate
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA.,Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Koichi Takahashi
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Tomoyuki Tanaka
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Noushin R Farnoud
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sherry Pierce
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph D Khoury
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey L Jorgensen
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P Patel
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Sa A Wang
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
12
|
Semchenkova A, Brilliantova V, Shelikhova L, Zhogov V, Illarionova O, Mikhailova E, Raykina E, Skorobogatova E, Novichkova G, Maschan A, Maschan M, Popov A. Chimerism evaluation in measurable residual disease‐suspected cells isolated by flow cell sorting as a reliable tool for measurable residual disease verification in acute leukemia patients after allogeneic hematopoietic stem cell transplantation. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:568-573. [DOI: 10.1002/cyto.b.21982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/27/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Alexandra Semchenkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Varvara Brilliantova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Larisa Shelikhova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Vladimir Zhogov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Olga Illarionova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Ekaterina Mikhailova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Elena Raykina
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | | | - Galina Novichkova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Alexey Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Michael Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| | - Alexander Popov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow Russia
| |
Collapse
|
13
|
Wang Y, Wang H, Wang W, Liu W, Liu N, Liu S, Lu Y. Prognostic value of platelet recovery degree before and after achieving minimal residual disease negative complete remission in acute myeloid leukemia patients. BMC Cancer 2020; 20:732. [PMID: 32758189 PMCID: PMC7409648 DOI: 10.1186/s12885-020-07222-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 07/27/2020] [Indexed: 12/19/2022] Open
Abstract
Background Risk stratification and prognosis prediction of acute myeloid leukemia (AML) are largely dependent on pre-treatment information. However, post-treatment data also provides much useful information. In this retrospective study, we explored whether the level of blood count recovery before and after the first minimal residual disease (MRD) negative complete remission (CR) is relevant to clinical outcomes of AML patients. Methods For each included patient, peripheral platelet counts were measured on the day before initial treatment (PLTpre), whereas platelet peak values (PLTpeak) were recorded after marrow recovery following the chemotherapy course inducing the first MRD-negative CR. The difference (DPLT) between these two values (DPLT = PLTpeak−PLTpre) was calculated. X-tile software was utilized to establish the optimal cut-point for DPLT, which was expected to distinguish CR patients with different clinical outcomes. A cross validation analysis was conducted to confirm the robustness of the established cut-point. The results were further tested by a Cox multivariate analysis. Results The optimal cut-point of DPLT was determined as 212 × 109/L. Patients in high DPLT group were observed to have a significantly better PFS (p = 0.016) and a better OS (without statistical significance, p = 0.106). Cox multivariate analysis showed that higher DPLT was associated with longer PFS (HR = 2.894, 95% CI: 1.320–6.345, p = 0.008) and longer OS (HR = 3.077, 95% CI: 1.130–8.376, p = 0.028). Conclusion Platelet recovery degree before and after achieving MRD-negative CR (DPLT) is a potential predictor of clinical outcomes in CR patients. Higher DPLT value is associated with longer PFS and OS. Our findings may help to develop simple methods for AML prognosis evaluation.
Collapse
Affiliation(s)
- Yang Wang
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfengdong Rd, Guangzhou, 510060, China.,State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Hua Wang
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfengdong Rd, Guangzhou, 510060, China.,State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Weida Wang
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfengdong Rd, Guangzhou, 510060, China.,State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Wenjian Liu
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfengdong Rd, Guangzhou, 510060, China.,State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Nawei Liu
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfengdong Rd, Guangzhou, 510060, China.,State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Shuang Liu
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfengdong Rd, Guangzhou, 510060, China.,State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yue Lu
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfengdong Rd, Guangzhou, 510060, China. .,State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| |
Collapse
|
14
|
Pettersson L, Chen Y, George AM, Rigo R, Lazarevic V, Juliusson G, Saal LH, Ehinger M. Subclonal patterns in follow-up of acute myeloid leukemia combining whole exome sequencing and ultrasensitive IBSAFE digital droplet analysis. Leuk Lymphoma 2020; 61:2168-2179. [PMID: 32425124 DOI: 10.1080/10428194.2020.1755855] [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: 12/17/2022]
Abstract
We studied mutation kinetics in ten relapsing and four non-relapsing patients with acute myeloid leukemia by whole exome sequencing at diagnosis to identify leukemia-specific mutations and monitored selected mutations at multiple time-points using IBSAFE droplet digital PCR. Five to nine selected mutations could identify and track leukemic clones prior to clinical relapse in 10/10 patients at the time-points where measurable residual disease was negative by multicolor flow cytometry. In the non-relapsing patients, the load of mutations gradually declined in response to different therapeutic strategies. Three distinct patterns of relapse were observed: (1) one or more different clones with all monitored mutations reappearing at relapse; (2) one or more separate clones of which one prevailed at relapse; and (3) persistent clonal hematopoiesis with high variant allele frequency and most mutations present at relapse. These pilot results demonstrate that IBSAFE analyses detect leukemic clones missed by flow cytometry with possible clinical implications.HighlightsThe IBSAFE ddPCR MRD method seems applicable on virtually all newly diagnosed AML patients and was more sensitive than flow cytometry.Monitoring a few mutations captured the kinetics of the evolving recurrent leukemia.NPM1-mutation alone may not be a reliable MRD-marker.
Collapse
Affiliation(s)
- Louise Pettersson
- Department of Pathology, Halland Hospital Halmstad, Region Halland, Halmstad, Sweden.,Department of Clinical Sciences, Division of Pathology, Lund University, Skane University Hospital, Lund, Sweden
| | - Yilun Chen
- Department of Clinical Sciences, Division of Oncology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Anthony M George
- Department of Clinical Sciences, Division of Oncology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Robert Rigo
- Department of Clinical Sciences, Division of Oncology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Vladimir Lazarevic
- Department of Hematology, Oncology and Radiation Physics, Lund University, Skane University Hospital, Lund, Sweden
| | - Gunnar Juliusson
- Department of Hematology, Oncology and Radiation Physics, Lund University, Skane University Hospital, Lund, Sweden.,Department of Laboratory Medicine, Stem Cell Center, Lund University, Skane University Hospital, Lund, Sweden
| | - Lao H Saal
- Department of Clinical Sciences, Division of Oncology, Faculty of Medicine, Lund University, Lund, Sweden.,Lund University Cancer Center, Medicon Village, Lund, Sweden
| | - Mats Ehinger
- Department of Clinical Sciences, Division of Pathology, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
15
|
M. Weerakoon-Ratnayake K, Vaidyanathan S, Larkey N, Dathathreya K, Hu M, Jose J, Mog S, August K, K. Godwin A, L. Hupert M, A. Witek M, A. Soper S. Microfluidic Device for On-Chip Immunophenotyping and Cytogenetic Analysis of Rare Biological Cells. Cells 2020; 9:E519. [PMID: 32102446 PMCID: PMC7072755 DOI: 10.3390/cells9020519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 01/09/2023] Open
Abstract
The role of circulating plasma cells (CPCs) and circulating leukemic cells (CLCs) as biomarkers for several blood cancers, such as multiple myeloma and leukemia, respectively, have recently been reported. These markers can be attractive due to the minimally invasive nature of their acquisition through a blood draw (i.e., liquid biopsy), negating the need for painful bone marrow biopsies. CPCs or CLCs can be used for cellular/molecular analyses as well, such as immunophenotyping or fluorescence in situ hybridization (FISH). FISH, which is typically carried out on slides involving complex workflows, becomes problematic when operating on CLCs or CPCs due to their relatively modest numbers. Here, we present a microfluidic device for characterizing CPCs and CLCs using immunofluorescence or FISH that have been enriched from peripheral blood using a different microfluidic device. The microfluidic possessed an array of cross-channels (2-4 µm in depth and width) that interconnected a series of input and output fluidic channels. Placing a cover plate over the device formed microtraps, the size of which was defined by the width and depth of the cross-channels. This microfluidic chip allowed for automation of immunofluorescence and FISH, requiring the use of small volumes of reagents, such as antibodies and probes, as compared to slide-based immunophenotyping and FISH. In addition, the device could secure FISH results in <4 h compared to 2-3 days for conventional FISH.
Collapse
Affiliation(s)
- Kumuditha M. Weerakoon-Ratnayake
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Swarnagowri Vaidyanathan
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
- Bioengineering, The University of Kansas, Lawrence, KS 66045, USA
| | - Nicholas Larkey
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Kavya Dathathreya
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Mengjia Hu
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Jilsha Jose
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Shalee Mog
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Keith August
- Children’s Mercy Hospital, Kansas City, MO 64108, USA;
| | - Andrew K. Godwin
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Mateusz L. Hupert
- Biofluidica Inc., BioFluidica Research Laboratory, Lawrence, KS 66047, USA
| | - Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Biofluidica Inc., BioFluidica Research Laboratory, Lawrence, KS 66047, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
16
|
Schuurhuis GJ, Ossenkoppele GJ, Kelder A, Cloos J. Measurable residual disease in acute myeloid leukemia using flow cytometry: approaches for harmonization/standardization. Expert Rev Hematol 2019; 11:921-935. [PMID: 30466339 DOI: 10.1080/17474086.2018.1549479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Measurable residual disease (MRD) in acute myeloid leukemia (AML) is a rapidly evolving area with many institutes embarking on it, both in academic and pharmaceutical settings. However, there is a multitude of approaches to design, perform, and report flow cytometric MRD. Together with the long-term experience needed, this makes flow cytometric MRD in AML nonstandardized and time-consuming. Areas covered: This paper briefly summarizes critical issues, like sample preparation and transport, markers and fluorochromes of choice, but in particular focuses on the main issues, which includes specificity and sensitivity, hereby providing a new model that may circumvent the main disadvantages of the present approaches. New approaches that may add to the value of flow cytometric MRD includes assessment of leukemia stem cells, MRD in peripheral blood, and approaches to use multidimensional image analysis. Expert commentary: MRD in AML requires standardization/harmonization on many aspects, for which the present paper offers possible guidelines.
Collapse
Affiliation(s)
- Gerrit J Schuurhuis
- a Department of Hematology , VU University Medical Center , Amsterdam , Netherlands
| | - Gert J Ossenkoppele
- a Department of Hematology , VU University Medical Center , Amsterdam , Netherlands
| | - Angèle Kelder
- a Department of Hematology , VU University Medical Center , Amsterdam , Netherlands
| | - Jacqueline Cloos
- a Department of Hematology , VU University Medical Center , Amsterdam , Netherlands
| |
Collapse
|
17
|
Selim A, Alvaro F, Cole CH, Fraser CJ, Mechinaud F, O'Brien TA, Shaw PJ, Tapp H, Teague L, Nivison-Smith I, Moore AS. Hematopoietic stem cell transplantation for children with acute myeloid leukemia in second remission: A report from the Australasian Bone Marrow Transplant Recipient Registry and the Australian and New Zealand Children's Haematology Oncology Group. Pediatr Blood Cancer 2019; 66:e27812. [PMID: 31111633 DOI: 10.1002/pbc.27812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/10/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Approximately one-third of children with acute myeloid leukemia (AML) relapse, requiring re-treatment and allogeneic hematopoietic stem cell transplantation (HSCT). Although achieving second complete remission (CR2) prior to HSCT is desirable, once CR2 is attained, it is unclear if there is any benefit from further chemotherapy prior to HSCT. Moreover, although pre-HSCT minimal residual disease (MRD) has prognostic value in acute lymphoblastic leukemia, the benefit of MRD reduction after achieving CR prior to HSCT is less clear for AML. PROCEDURE To address these questions, we analyzed data from pediatric transplant centers in Australia and New Zealand concerning relapsed childhood AML cases occurring between 1998 and 2013. Given the retrospective nature of our analysis and assay data available, we analyzed patients on the basis of measurable residual disease (MeRD) by any methodology, rather than MRD in the conventional sense. RESULTS We observed improved overall survival (OS) in children receiving two chemotherapy cycles, compared to one cycle or three or more cycles pre-HSCT. Improved OS with two cycles remained significant for patients without MeRD after cycle 1. CONCLUSIONS These data suggest that a second chemotherapy cycle pre-HSCT may improve survival by lowering disease burden. Prospective trials assessing strategies to reduce pre-HSCT MRD in relapsed childhood AML are warranted.
Collapse
Affiliation(s)
- Adrian Selim
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | | | - Catherine H Cole
- Department of Haematology/Oncology, Princess Margaret Hospital for Children, Perth, Australia
| | - Chris J Fraser
- Oncology Services Group, Queensland Children's Hospital, Brisbane, Australia
| | | | - Tracey A O'Brien
- Kids' Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Peter J Shaw
- Oncology Unit, The Children's Hospital at Westmead, Sydney, Australia
| | - Heather Tapp
- Department of Clinical Haematology/Oncology, Women's and Children's Hospital, Adelaide, Australia
| | | | - Ian Nivison-Smith
- Australasian Bone Marrow Transplant Recipient Registry, Sydney, Australia
| | - Andrew S Moore
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Oncology Services Group, Queensland Children's Hospital, Brisbane, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
18
|
Camburn AE, Petrasich M, Ruskova A, Chan G. Myeloblasts in normal bone marrows expressing leukaemia-associated immunophenotypes. Pathology 2019; 51:502-506. [PMID: 31262563 DOI: 10.1016/j.pathol.2019.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 03/17/2019] [Accepted: 03/21/2019] [Indexed: 01/07/2023]
Abstract
Measurable residual disease (MRD) status of patients undergoing treatment for acute myeloid leukaemia (AML) is important for prognosis and guides treatment. Multicolour flow cytometry (MCF) is a sensitive MRD method. The current approach relies on identification of blasts expressing leukaemia-associated immunophenotypes (LAIP) or by blasts expressing aberrant differentiation/maturation profiles compared to that seen in normal haematopoietic precursor cells at follow-up, i.e., different from normal (DFN). However, expression of LAIP on normal myeloblasts affects the specificity of the result, and the understanding of what is normal is important. Limited published data are currently available. We report findings from 14 normal adult bone marrows. MCF was performed on the residual normal marrow specimens from 14 adults. Expression of CD15, CD11b, CD7, CD4, and CD56 on CD34+ myeloblasts was assessed. Analysis of samples was performed using 4-colour flow cytometry which was the methodology used when this work was done, and is still being used in many clinical flow laboratories worldwide. LAIP is defined by lineage infidelity or asynchronous expression of differentiation markers. The cases of normal myeloblasts with LAIP involving the markers used and above the cut-off levels for MRD detection (0.01%) varies between 43% and 100%, limiting the specificity of the results for MRD. Even if the threshold is raised to 0.1%, there will still be false positive cases using aberrant CD15 or CD7. Our work provided useful information for AML MRD determination in our laboratory. A collaborative database of LAIP on normal myeloblasts using standardised analysis should be useful to determine the optimal diagnostic cut-off for AML MRD using LAIP.
Collapse
Affiliation(s)
| | | | - Anna Ruskova
- LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - George Chan
- LabPlus, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
19
|
Sui J, Chen Q, Zhang Y, Sheng Y, Wu J, Li J, Weng X, Chen B. Identifying leukemia-associated immunophenotype-based individualized minimal residual disease in acute myeloid leukemia and its prognostic significance. Am J Hematol 2019; 94:528-538. [PMID: 30734356 DOI: 10.1002/ajh.25431] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 12/25/2022]
Abstract
Based on the leukemia-associated immunophenotypes (LAIPs), minimal residual disease (MRD) related to the outcome can be detected by multiparameter flow cytometry in acute myeloid leukemia (AML) patients. Although 0.1% was commonly used as a cutoff value, measurable MRD or MRD level below 0.1% has also been associated with prognostic significance and more sensitive thresholds (<0.1%) are required for improving AML prognosis prediction. In this study, 292 adult patients diagnosed with AML (non-M3) were enrolled, 36 kinds of LAIPs were identified, and the baseline expression levels in normal or regenerating bone marrows of each kind of LAIP were established, which ranged from <2.00 × 10-5 to 5.71 × 10-4 . The baseline level of each LAIP was considered as the individual threshold for MRD assessment. MRD statuses stratified by 0.1% and individual threshold were termed as 0.1%-MRD and individual-MRD, respectively. The patients of individual-MRDneg showed significantly better survival compared with those of 0.1%-MRDneg /individual-MRDpos or 0.1%-MRDpos . Multivariate analysis showed that when time points of complete remission, post the first and second consolidation courses, were considered, only individual-MRD post second consolidation presented independent prognostic value. Notably, in patients of cytogenetic/molecular low-risk (LR) or intermediate-risk (IR), the individual-MRD status could identify the patients with significantly different outcomes, while 0.1%-MRD status could not. Furthermore, among the patients of the LR or IR group which received chemotherapy only, those with individual-MRDneg status presented favorable survival, which was comparable with that of the patients accepted allogeneic hematopoietic stem cell transplantation (ASCT). This approach is useful in the selection of an ASCT strategy for clinical practice.
Collapse
Affiliation(s)
- Jing‐Ni Sui
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Qiu‐Sheng Chen
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Yun‐Xiang Zhang
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Yan Sheng
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Jing Wu
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Jun‐Min Li
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Xiang‐Qin Weng
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Bing Chen
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| |
Collapse
|
20
|
Ehinger M, Pettersson L. Measurable residual disease testing for personalized treatment of acute myeloid leukemia. APMIS 2019; 127:337-351. [PMID: 30919505 DOI: 10.1111/apm.12926] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022]
Abstract
This review summarizes - with the practicing hematologist in mind - the methods used to determine measurable residual disease (MRD) in everyday practice with some future perspectives, and the current knowledge about the prognostic impact of MRD on outcome in acute myeloid leukemia (AML), excluding acute promyelocytic leukemia. Possible implications for choice of MRD method, timing of MRD monitoring, and guidance of therapy are discussed in general and in some detail for certain types of leukemia with specific molecular markers to monitor, including core binding factor (CBF)-leukemias and NPM1-mutated leukemias.
Collapse
Affiliation(s)
- Mats Ehinger
- Department of Clinical Sciences, Pathology, Skane University Hospital, Lund University, Lund, Sweden
| | - Louise Pettersson
- Department of Pathology, Halland Hospital Halmstad, Region Halland, Halmstad, Sweden.,Faculty of Medicine, Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
21
|
Veltri L, Rezvani K, Oran B, Mehta R, Rondon G, Kebriaei P, Popat U, Nieto Y, Hosing C, Qazilbash M, Khouri I, Shpall E, Champlin R, Marin D. Allotransplants for Patients 65 Years or Older with High-Risk Acute Myeloid Leukemia. Biol Blood Marrow Transplant 2018; 25:505-514. [PMID: 30308325 DOI: 10.1016/j.bbmt.2018.09.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/27/2018] [Indexed: 01/09/2023]
Abstract
The outcome of persons > 65 years with acute myeloid leukemia (AML) is poor. A transplant from an HLA-identical sibling or an HLA-matched unrelated donor can cure some of these patients but is associated with a substantial transplant-related mortality and a high relapse risk. We analyzed 185 subjects > 65 years with high-risk AML receiving conventional (n = 42) or reduced-intensity (n = 143) pretransplant conditioning and a transplant from an HLA-identical sibling (n = 66) or a 10/10 loci HLA-matched unrelated donor (n = 119). Two-year survival was 37%. Subjects with serious adverse events during before chemotherapy for their leukemia had a poor outcome after stem cell transplantation. Patients who had active leukemia or measurable residual disease (MRD) before transplantation had a worse outcome. Delayed hematologic recovery after induction or consolidation chemotherapy, high-risk AML genetics, donor-recipient HLA-DRβ3/4/5-DP mismatches, and history of cardiovascular disease were also correlated with survival in multivariate analyses. The 57 MRD-negative patients with few other adverse prognostic factors had an excellent outcome (2-year overall survival, 76%), whereas the 58 patients with detectable leukemia and more than 1 other additional factor fared poorly (2-year overall survival, 8%). These data indicate it is possible to identify persons > 65 years with high-risk AML likely to benefit from an allotransplant. Validation of this prediction is needed.
Collapse
Affiliation(s)
- Lauren Veltri
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Betul Oran
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Rohtesh Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Muzaffar Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Issa Khouri
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, Texas.
| |
Collapse
|
22
|
Methods and role of minimal residual disease after stem cell transplantation. Bone Marrow Transplant 2018; 54:681-690. [PMID: 30116018 DOI: 10.1038/s41409-018-0307-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/28/2018] [Accepted: 06/13/2018] [Indexed: 11/08/2022]
Abstract
Relapse is the major cause of treatment failure after stem cell transplantation. Despite the fact that relapses occurred even if transplantation was performed in complete remission, it is obvious that minimal residual disease is present though not morphologically evident. Since adaptive immunotherapy by donor lymphocyte infusion or other novel cell therapies as well as less toxic drugs, which can be used after transplantation, the detection of minimal residual disease (MRD) has become a clinical important variable for outcome. Besides the increasing options to treat MRD, the most advanced technologies currently allow to detect residual malignant cells with a sensitivity of 10-5 to 10-6.Under the patronage of the European Society for Blood and Marrow Transplantation (EBMT) and the American Society for Blood and Marrow Transplantation (ASBMT) the 3rd workshop was held on 4/5 November 2016 in Hamburg/Germany, with the aim to present an up-to-date status of epidemiology and biology of relapse and to summarize the currently available options to prevent and treat post-transplant relapse. Here the current methods and role of minimal residual disease for myeloid and lymphoid malignancies are summarized.
Collapse
|
23
|
Early Post-Transplant Minimal Residual Disease Assessment Improves Risk Stratification in Acute Myeloid Leukemia. Biol Blood Marrow Transplant 2018; 24:1514-1520. [DOI: 10.1016/j.bbmt.2018.02.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/06/2018] [Indexed: 11/21/2022]
|
24
|
Selim AG, Moore AS. Molecular Minimal Residual Disease Monitoring in Acute Myeloid Leukemia. J Mol Diagn 2018; 20:389-397. [DOI: 10.1016/j.jmoldx.2018.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/22/2018] [Accepted: 03/27/2018] [Indexed: 01/22/2023] Open
|
25
|
Buccisano F, Dillon R, Freeman SD, Venditti A. Role of Minimal (Measurable) Residual Disease Assessment in Older Patients with Acute Myeloid Leukemia. Cancers (Basel) 2018; 10:cancers10070215. [PMID: 29949858 PMCID: PMC6070940 DOI: 10.3390/cancers10070215] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/18/2018] [Accepted: 06/18/2018] [Indexed: 12/22/2022] Open
Abstract
Minimal (or measurable) residual (MRD) disease provides a biomarker of response quality for which there is robust validation in the context of modern intensive treatment for younger patients with Acute Myeloid Leukemia (AML). Nevertheless, it remains a relatively unexplored area in older patients with AML. The lack of progress in this field can be attributed to two main reasons. First, physicians have a general reluctance to submitting older adults to intensive chemotherapy due to their frailty and to the unfavourable biological disease profile predicting a poor outcome following conventional chemotherapy. Second, with the increasing use of low-intensity therapies (i.e., hypomethylating agents) differing from conventional drugs in mechanism of action and dynamics of response, there has been concomitant skepticism that these schedules can produce deep hematological responses. Furthermore, age dependent differences in disease biology also contribute to uncertainty on the prognostic/predictive impact in older adults of certain genetic abnormalities including those validated for MRD monitoring in younger patients. This review examines the evidence for the role of MRD as a prognosticator in older AML, together with the possible pitfalls of MRD evaluation in older age.
Collapse
Affiliation(s)
- Francesco Buccisano
- Hematology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London SE1 9RT, UK.
| | - Sylvie D Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK.
| | - Adriano Venditti
- Hematology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
| |
Collapse
|
26
|
Shumilov E, Flach J, Kohlmann A, Banz Y, Bonadies N, Fiedler M, Pabst T, Bacher U. Current status and trends in the diagnostics of AML and MDS. Blood Rev 2018; 32:508-519. [PMID: 29728319 DOI: 10.1016/j.blre.2018.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 01/01/2023]
Abstract
Diagnostics of acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) have recently been experiencing extensive modifications regarding the incorporation of next-generation sequencing (NGS) strategies into established diagnostic algorithms, classification and risk stratification systems, and minimal residual disease (MRD) detection. Considering the increasing arsenal of targeted therapies (e.g. FLT3 or IDH1/IDH2 inhibitors) for AML, timely and comprehensive molecular mutation screening has arrived in daily practice. Next-generation flow strategies allow for immunophenotypic minimal residual disease (MRD) monitoring with very high sensitivity. At the same time, standard diagnostic tools such as cytomorphology or conventional cytogenetics remain cornerstones for the diagnostic workup of myeloid malignancies. Herein, we summarize the most recent advances and new trends for the diagnostics of AML and MDS, discuss the difficulties, which accompany the integration of these new methods and their results into daily routine, and aim to define the role hemato-oncologists may play in this new diagnostic era.
Collapse
Affiliation(s)
- Evgenii Shumilov
- Department of Hematology and Medical Oncology, University Medicine Göttingen (UMG), Göttingen, Germany
| | - Johanna Flach
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Alexander Kohlmann
- Precision Medicine and Genomics, Innovative Medicines and Early Development, AstraZeneca, Cambridge, UK
| | - Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Nicolas Bonadies
- University Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, Inselspital, Bern, Bern University Hospital, University of Bern, Switzerland
| | - Martin Fiedler
- Center of Laboratory Medicine (ZLM)/University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland.
| | - Ulrike Bacher
- University Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, Bern, Switzerland; Center of Laboratory Medicine (ZLM)/University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland.
| |
Collapse
|
27
|
Happle C, Meyer-Decking L, Dreier A, Wetzke M, Gläsener S, Grychtol R, Braubach P, Jablonka A, Haid S, Jirmo AC, Habener A, Skuljec J, Hansen G. Improved protocol for simultaneous analysis of leukocyte subsets and epithelial cells from murine and human lung. Exp Lung Res 2018; 44:127-136. [PMID: 29677457 DOI: 10.1080/01902148.2018.1432721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE To study and isolate lung cells by flow cytometry, enzymatic digestion and generation of single cell suspensions is required. This significantly influences expression of cellular epitopes and protocols need to be adapted for the best isolation and subsequent analysis of specific cellular subsets. MATERIALS AND METHODS We optimized protocols for the simultaneous isolation and characterization of specific human and murine lung cell types. For alveolar epithelial cells (AEC), a primarily dispase based digestion method and for leukocytes, a primarily collagenase based technique was adapted. Protocols were applied in parallel in either single experimental mice or human lung specimens. RESULTS Optimized dispase/DNase digestion yielded a high percentage of Epcam+CD45-CD31- AEC as assessed by flow cytometry. Epcam+CD45-CD3-CD11b-CD11c-CD16/32-CD19-CD31-F4/80- AEC were readily sortable with high purity and typical morphology and function upon in vitro stimulation with lipopolysaccharide or respiratory-syncytial-virus (RSV) infection. To analyze lung leukocytes, specimens were digested with an adapted collagenase/DNase protocol yielding high percentages of viable leukocytes with typical morphology, function, and preserved subset specific leukocyte markers. Both protocols could be applied simultaneously in a single experimental mouse post mortem. Application of both digestion methods in primary human lung specimens yielded similar results with high proportions of Epcam+CD45- human AEC after dispase/DNase digestion and preservation of human T cell epitopes after collagenase/DNase digestion. CONCLUSION The here described protocols were optimized for the simple and efficient isolation of murine and human lung cells. In contrast to previously described techniques, they permit simultaneous in-depth characterization of pulmonary epithelial cells and leukocyte subsets such as T helper, cytotoxic T, and B cells from one sample. As such, they may help to comprehensively and sustainably characterize murine and human lung specimens and facilitate studies on the role of lung immune cells in different respiratory pathologies.
Collapse
Affiliation(s)
- Christine Happle
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Lena Meyer-Decking
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany
| | - Anika Dreier
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Martin Wetzke
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany
| | - Stephanie Gläsener
- b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Ruth Grychtol
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Peter Braubach
- b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany.,c Department of Pathology , Hannover Medical School , Hannover , Germany
| | - Alexandra Jablonka
- d Department of Clinical Immunology and Rheumatology , Hannover Medical School , Hannover , Germany.,e German Center for Infection Research (DZIF) , Standort Hannover-Braunschweig , Germany
| | - Sibylle Haid
- f Experimental Virology and TWINCORE , a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research , Hannover , Germany
| | - Adan Chari Jirmo
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Anika Habener
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| | - Jelena Skuljec
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany
| | - Gesine Hansen
- a Department of Pediatrics, Allergology, and Neonatology , Hannover Medical School , Hannover , Germany.,b German Center for Lung Research , Biomedical Research in End Stage and Obstructive Lung Disease/ BREATH Hannover , Germany
| |
Collapse
|
28
|
[Expert consensus on minimal residual disease detection of acute leukemia and plasma cell neoplasms by multi-parameter flow cytometry]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 38:1001-1011. [PMID: 29365391 PMCID: PMC7342185 DOI: 10.3760/cma.j.issn.0253-2727.2017.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Indexed: 11/09/2022]
|
29
|
Daver N, Kantarjian H, Garcia-Manero G, Jabbour E, Borthakur G, Brandt M, Pierce S, Vaughan K, Ning J, Nogueras González GM, Patel K, Jorgensen J, Pemmaraju N, Kadia T, Konopleva M, Andreeff M, DiNardo C, Cortes J, Ward R, Craig A, Ravandi F. Vosaroxin in combination with decitabine in newly diagnosed older patients with acute myeloid leukemia or high-risk myelodysplastic syndrome. Haematologica 2017; 102:1709-1717. [PMID: 28729302 PMCID: PMC5622855 DOI: 10.3324/haematol.2017.168732] [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: 03/17/2017] [Accepted: 07/12/2017] [Indexed: 11/09/2022] Open
Abstract
Vosaroxin is an anti-cancer quinolone-derived DNA topoisomerase II inhibitor. We investigated vosaroxin with decitabine in patients ≥60 years of age with newly diagnosed acute myeloid leukemia (n=58) or myelodysplastic syndrome (≥10% blasts) (n=7) in a phase II non-randomized trial. The initial 22 patients received vosaroxin 90 mg/m2 on days 1 and 4 with decitabine 20 mg/m2 on days 1–5 every 4–6 weeks for up to seven cycles. Due to a high incidence of mucositis the subsequent 43 patients were given vosaroxin 70 mg/m2 on days 1 and 4. These 65 patients, with a median age of 69 years (range, 60–78), some of whom with secondary leukemia (22%), adverse karyotype (35%), or TP53 mutation (20%), are evaluable. The overall response rate was 74% including complete remission in 31 (48%), complete remission with incomplete platelet recovery in 11 (17%), and complete remission with incomplete count recovery in six (9%). The median number of cycles to response was one (range, 1–4). Grade 3/4 mucositis was noted in 17% of all patients. The 70 mg/m2 induction dose of vosaroxin was associated with similar rates of overall response (74% versus 73%) and complete remission (51% versus 41%, P=0.44), reduced incidence of mucositis (30% versus 59%, P=0.02), reduced 8-week mortality (9% versus 23%; P=0.14), and improved median overall survival (14.6 months versus 5.5 months, P=0.007). Minimal residual disease-negative status by multiparametric flow-cytometry at response (± 3 months) was achieved in 21 of 39 (54%) evaluable responders and was associated with better median overall survival (34.0 months versus 8.3 months, P=0.023). In conclusion, the combination of vosaroxin with decitabine is effective and well tolerated at a dose of 70 mg/m2 and warrants randomized prospective evaluation. ClinicalTrials.gov: NCT01893320
Collapse
Affiliation(s)
- Naval Daver
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Mark Brandt
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Sherry Pierce
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kenneth Vaughan
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jing Ning
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | - Keyur Patel
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jeffery Jorgensen
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Michael Andreeff
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Courtney DiNardo
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jorge Cortes
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Renee Ward
- Sunesis Pharmaceuticals Inc., South San Francisco, CA, USA
| | - Adam Craig
- Sunesis Pharmaceuticals Inc., South San Francisco, CA, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
30
|
Cruz NM, Mencia-Trinchant N, Hassane DC, Guzman ML. Minimal residual disease in acute myelogenous leukemia. Int J Lab Hematol 2017; 39 Suppl 1:53-60. [PMID: 28447422 DOI: 10.1111/ijlh.12670] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/03/2017] [Indexed: 12/21/2022]
Abstract
Treatment of acute myelogenous leukemia (AML) over the past four decades remains mostly unchanged and the prognosis for the majority of patients remains poor. Most of the significant advances that have been observed are in defining cytogenetic abnormalities, as well as the genetic and epigenetic profiles of AML patients. While new cytogenetic and genetic aberrations such as the FLT3-ITD and NPM1 mutations are able to guide prognosis for the majority of patients with AML, outcomes are still dismal and relapse rates remain high. It is thought that relapse in AML is in part driven by minimal residual disease (MRD) that remains in the patient following treatment. Thus, there is a need for sensitive and objective methodology for MRD detection. Methodologies such as multiparameter flow cytometry (MFC), quantitative real-time polymerase chain reaction (RQ-PCR), digital PCR (dPCR), or next-generation sequencing (NGS) are being employed to evaluate their utility in MRD assessment. In this review, we will provide an overview of AML and the clinical utility of MRD measurement. We will discuss optimal timing to MRD measurement, the different approaches that are available, and efforts in the standardization across laboratories.
Collapse
Affiliation(s)
- N M Cruz
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - N Mencia-Trinchant
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - D C Hassane
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - M L Guzman
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
31
|
Williams-Voorbeijtel D, Sanchez F, Roth CG. Aligning the flow cytometric evaluation with the diagnostic need: an evidence-based approach. J Clin Pathol 2017; 70:740-744. [DOI: 10.1136/jclinpath-2016-204316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 01/31/2023]
Abstract
AimsElimination of non-value added testing without compromising high-quality clinical care is an important mandate for laboratories in a value-based reimbursement system. The goal of this study was to determine the optimal combination of flow cytometric markers for a screening approach that balances efficiency and accuracy.MethodsAn audit over 9 months of flow cytometric testing was performed, including rereview of all dot plots from positive cases.ResultsOf the 807 cases in which leukaemia/lymphoma testing was performed, 23 were non-diagnostic and 189 represented bronchoalveolar lavage specimens. Of the remaining 595 cases, 137 (23%) were positive for an abnormal haematolymphoid population. Review of the positive cases identified minimum requirements for a screening tube as well as analysis strategies to overcome the diagnostic pitfalls noted. It is estimated that 38% fewer antibodies would be used in a screening approach, representing an opportunity for significant cost savings.ConclusionsWe provide a framework for developing an evidence-based screening combination for cost-effective characterisation of haematolymphoid malignancies, promoting adoption of ‘just-in-time’ testing systems that tailor the evaluation to the diagnostic need.
Collapse
|
32
|
Han L, Jorgensen JL, Brooks C, Shi C, Zhang Q, Nogueras González GM, Cavazos A, Pan R, Mu H, Wang SA, Zhou J, Ai-Atrash G, Ciurea SO, Rettig M, DiPersio JF, Cortes J, Huang X, Kantarjian HM, Andreeff M, Ravandi F, Konopleva M. Antileukemia Efficacy and Mechanisms of Action of SL-101, a Novel Anti-CD123 Antibody Conjugate, in Acute Myeloid Leukemia. Clin Cancer Res 2017; 23:3385-3395. [PMID: 28096272 DOI: 10.1158/1078-0432.ccr-16-1904] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/13/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022]
Abstract
Purpose: The persistence of leukemia stem cells (LSC)-containing cells after induction therapy may contribute to minimal residual disease (MRD) and relapse in acute myeloid leukemia (AML). We investigated the clinical relevance of CD34+CD123+ LSC-containing cells and antileukemia potency of a novel antibody conjugate SL-101 in targeting CD123+ LSCs.Experimental Methods and Results: In a retrospective study on 86 newly diagnosed AML patients, we demonstrated that a higher proportion of CD34+CD123+ LSC-containing cells in remission was associated with persistent MRD and predicted shorter relapse-free survival in patients with poor-risk cytogenetics. Using flow cytometry, we explored the potential benefit of therapeutic targeting of CD34+CD38-CD123+ cells by SL-101, a novel antibody conjugate comprising an anti-CD123 single-chain Fv fused to Pseudomonas exotoxin A The antileukemia potency of SL-101 was determined by the expression levels of CD123 antigen in a panel of AML cell lines. Colony-forming assay established that SL-101 strongly and selectively suppressed the function of leukemic progenitors while sparing normal counterparts. The internalization, protein synthesis inhibition, and flow cytometry assays revealed the mechanisms underlying the cytotoxic activities of SL-101 involved rapid and efficient internalization of antibody, sustained inhibition of protein synthesis, induction of apoptosis, and blockade of IL3-induced p-STAT5 and p-AKT signaling pathways. In a patient-derived xenograft model using NSG mice, the repopulating capacity of LSCs pretreated with SL-101 in vitro was significantly impaired.Conclusions: Our data define the mechanisms by which SL-101 targets AML and warrant further investigation of the clinical application of SL-101 and other CD123-targeting strategies in AML. Clin Cancer Res; 23(13); 3385-95. ©2017 AACR.
Collapse
Affiliation(s)
- Lina Han
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jeffrey L Jorgensen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Ce Shi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Antonio Cavazos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rongqing Pan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Mu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sa A Wang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jin Zhou
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gheath Ai-Atrash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan O Ciurea
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mike Rettig
- Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Jorge Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
33
|
Dehghan-Nayeri N, Rezaei-Tavirani M, Omrani MD, Gharehbaghian A, Goudarzi Pour K, Eshghi P. Identification of potential predictive markers of dexamethasone resistance in childhood acute lymphoblastic leukemia. J Cell Commun Signal 2016; 11:137-145. [PMID: 27778231 DOI: 10.1007/s12079-016-0357-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/27/2016] [Indexed: 12/30/2022] Open
Abstract
Response to dexamethasone (DEXA), as a hallmark drug in the treatment of childhood acute lymphoblastic leukemia (ALL), is one of the pivotal prognostic factors in the prediction of outcome in ALL. Identification of predictive markers of chemoresistance is beneficial to selecting of the best therapeutic protocol with the lowest effect adverse. Hence, we aimed to find drug targets using the 2DE/MS proteomics study of a DEXA-resistant cell line (REH) as a model for poor DEXA responding patients before and after drug treatment. Using the proteomic methods, three differentially expressed proteins were detected, including voltage dependent anion channel 1 (VDAC1), sorting Nexin 3 (SNX3), and prefoldin subunit 6 (PFDN6). We observed low expression of three proteins after DEXA treatment in REH cells. We subsequently verified low expression of resulted proteins at the mRNA level using the quantitative PCR method. These proteins are promising proteins because of their important roles in drug resistance and regulation of apoptosis (VDAC1), protein trafficking (SNX3), and protein folding (PFDN6). Additionally, mRNA expression level of these proteins was assessed in 17 bone marrow samples from children with newly diagnosed ALL and 7 non-cancerous samples as controls. The results indicated that independent of the molecular subtypes of leukemia, mRNA expression of VDAC1, SNX3, and PFDN6 decreased in ALL samples compared with non-cancerous samples particularly in VDAC1 (p < 0.001). Additionally, mRNA expression of three proteins was also declined in high-risk samples compared with standard risk cases. These results demonstrated diagnostic and prognostic value of these proteins in childhood ALL. Furthermore, investigation of protein-protein interaction using STRING database indicated that these proteins involved in the signaling pathway of NR3C1 as dexamethasone target. In conclusion, our proteomic study in DEXA resistant leukemic cells revealed VDAC1, SNX3, and PFDN6 are promising proteins that might serve as potential biomarkers of prognosis and chemotherapy in childhood ALL.
Collapse
Affiliation(s)
- Nasrin Dehghan-Nayeri
- Proteomics Research Center, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Gharehbaghian
- Department of Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Kourosh Goudarzi Pour
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Peyman Eshghi
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Ravandi F, Jorgensen J, Borthakur G, Jabbour E, Kadia T, Pierce S, Brandt M, Wang S, Konoplev S, Wang X, Huang X, Daver N, DiNardo C, Andreeff M, Konopleva M, Estrov Z, Garcia-Manero G, Cortes J, Kantarjian H. Persistence of minimal residual disease assessed by multiparameter flow cytometry is highly prognostic in younger patients with acute myeloid leukemia. Cancer 2016; 123:426-435. [PMID: 27657543 DOI: 10.1002/cncr.30361] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/04/2016] [Accepted: 08/25/2016] [Indexed: 01/22/2023]
Abstract
BACKGROUND Predicting outcomes for patients with acute myeloid leukemia (AML) on the basis of pretreatment predictors has been the cornerstone of management. Posttreatment prognostic factors are increasingly being evaluated. METHODS Among 280 younger patients who were treated with intermediate-dose cytarabine (total ≥ 5 g/m2 ) and idarubicin-based induction chemotherapy and achieved remission, 186 were assessed for minimal residual disease (MRD) with an 8-color multiparameter flow cytometry panel performed on bone marrow specimens with a sensitivity of 0.1% or higher. RESULTS One hundred sixty-six patients had samples available 1 to 2 months after induction at the time of complete remission (CR), and 79% became negative for MRD, with an MRD-negative status associated with an improvement in relapse-free survival (RFS; P = .0002) and overall survival (OS; P = .0002). One hundred sixteen were evaluated for their MRD status during consolidation, and 86% were negative, with an MRD-negative status associated with a significant improvement in RFS (P < .0001) and OS (P < .0001). Sixty-nine patients were evaluated for their MRD status after completion of all therapy, and 84% were negative, with an MRD-negative status associated with an improvement in RFS (P < .0001) and OS (P < .0001). In a multivariate analysis including age, cytogenetics, response (CR vs CR with incomplete platelet recovery/incomplete blood count recovery), and MRD, achieving an MRD-negative status was the most important independent predictor of RFS and OS at response (P = .008 and P = .0008, respectively), during consolidation (P < .0001 for both), and at the completion of therapy (P < .0001 and P = .002, respectively). CONCLUSIONS Achieving an MRD-negative status according to multiparameter flow cytometry is associated with a highly significant improvement in the outcomes of younger patients with AML receiving cytosine arabinoside plus idarubicin-based induction and consolidation regimens. Cancer 2017;123:426-435. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey Jorgensen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark Brandt
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sa Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sergej Konoplev
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Courtney DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zeev Estrov
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Jorge Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
35
|
Oran B, Jorgensen JL, Marin D, Wang S, Ahmed S, Alousi AM, Andersson BS, Bashir Q, Bassett R, Lyons G, Chen J, Rezvani K, Popat U, Kebriaei P, Patel K, Rondon G, Shpall EJ, Champlin RE. Pre-transplantation minimal residual disease with cytogenetic and molecular diagnostic features improves risk stratification in acute myeloid leukemia. Haematologica 2016; 102:110-117. [PMID: 27540139 DOI: 10.3324/haematol.2016.144253] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/18/2016] [Indexed: 01/02/2023] Open
Abstract
Our aim was to improve outcome prediction after allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia by combining cytogenetic and molecular data at diagnosis with minimal residual disease assessment by multicolor flow-cytometry at transplantation. Patients with acute myeloid leukemia in first complete remission in whom minimal residual disease was assessed at transplantation were included and categorized according to the European LeukemiaNet classification. The primary outcome was 1-year relapse incidence after transplantation. Of 152 patients eligible, 48 had minimal residual disease at the time of their transplant. Minimal residual disease-positive patients were older, required more therapy to achieve first remission, were more likely to have incomplete recovery of blood counts and had more adverse risk features by cytogenetics. Relapse incidence at 1 year was higher in patients with minimal residual disease (32.6% versus 14.4%, P=0.002). Leukemia-free survival (43.6% versus 64%, P=0.007) and overall survival (48.8% versus 66.9%, P=0.008) rates were also inferior in patients with minimal residual disease. In multivariable analysis, minimal residual disease status at transplantation independently predicted 1-year relapse incidence, identifying a subgroup of intermediate-risk patients, according to the European LeukemiaNet classification, with a particularly poor outcome. Assessment of minimal residual disease at transplantation in combination with cytogenetic and molecular findings provides powerful independent prognostic information in acute myeloid leukemia, lending support to the incorporation of minimal residual disease detection to refine risk stratification and develop a more individualized approach during hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Betül Oran
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeff L Jorgensen
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sa Wang
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sairah Ahmed
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amin M Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roland Bassett
- Department Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Genevieve Lyons
- Department Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julianne Chen
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katy Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur Patel
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
36
|
Estey E. Acute myeloid leukemia: 2016 Update on risk-stratification and management. Am J Hematol 2016; 91:824-46. [PMID: 27417880 DOI: 10.1002/ajh.24439] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 11/09/2022]
Abstract
Evidence suggest that even patients aged 70 or above benefit from specific AML therapy. The fundamental decision in AML then becomes whether to recommend standard or investigational treatment. This decision must rest on the likely outcome of standard treatment. Hence we review factors that predict treatment related mortality and resistance to therapy, the latter the principal cause of failure even in patients aged 70 or above. We emphasize the limitations of prediction of resistance based only on pre- treatment factors and stress the need to incorporate post-treatment factors, for example indicators of minimal residual disease. We review various newer therapeutic options and considerations that underlie the decision to recommend allogeneic hematopoietic cell transplant. Am. J. Hematol. 91:825-846, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Elihu Estey
- Division of Hematology, University of Washington School of Medicine, Seattle, WA, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
37
|
Challenges in the harmonization of immune monitoring studies and trial design for cell-based therapies in the context of hematopoietic cell transplantation for pediatric cancer patients. Cytotherapy 2016; 17:1667-74. [PMID: 26589751 DOI: 10.1016/j.jcyt.2015.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/15/2015] [Accepted: 09/21/2015] [Indexed: 12/16/2022]
Abstract
Clinical trials aimed at improving results of hematopoietic cell transplantation (HCT) by adjuvant cell-based interventions in children have been limited by small numbers and pediatric-specific features. The need for a larger number of pediatric HCT centers to participate in trials has resulted in a demand for harmonization of disease-specific clinical trials and immune-monitoring. Thus far, most phase I/II trials select different end points evaluated at disparate time points, making inter-study comparisons difficult and, sometimes, impossible. In this review, we discuss the various aspects that are important to consider for harmonizing clinical trial design as well as the critical elements for standardized (immune)-monitoring protocols in cell-based intervention trials in the context of HCT. Comparison data from trials applying harmonized trial design will lead to optimized immunotherapeutic treatment protocols to maximize clinical efficacy while minimizing toxicity.
Collapse
|
38
|
Pettersson L, Levéen P, Axler O, Dvorakova D, Juliusson G, Ehinger M. Improved minimal residual disease detection by targeted quantitative polymerase chain reaction inNucleophosmin 1type a mutated acute myeloid leukemia. Genes Chromosomes Cancer 2016; 55:750-66. [DOI: 10.1002/gcc.22375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/25/2022] Open
Affiliation(s)
| | - Per Levéen
- Department of Pathology; University and Regional Laboratories, Skåne University Hospital; Lund Sweden
| | - Olof Axler
- Department of Pathology; University and Regional Laboratories, Skåne University Hospital; Lund Sweden
| | - Dana Dvorakova
- Department of Internal Medicine-Hematology and Oncology; Center of Molecular Biology and Gene Therapy, Masaryk University and University Hospital Brno; Brno Czech Republic
| | - Gunnar Juliusson
- Department of Hematology; Skåne University Hospital; Lund Sweden
| | - Mats Ehinger
- Department of Pathology; University and Regional Laboratories, Skåne University Hospital; Lund Sweden
| |
Collapse
|
39
|
Ouyang J, Goswami M, Peng J, Zuo Z, Daver N, Borthakur G, Tang G, Medeiros LJ, Jorgensen JL, Ravandi F, Wang SA. Comparison of Multiparameter Flow Cytometry Immunophenotypic Analysis and Quantitative RT-PCR for the Detection of Minimal Residual Disease of Core Binding Factor Acute Myeloid Leukemia. Am J Clin Pathol 2016; 145:769-77. [PMID: 27298396 DOI: 10.1093/ajcp/aqw038] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To examine the value of minimal residual disease (MRD) by multiparameter flow cytometry (MFC) in core binding factor (CBF) acute myeloid leukemia (AML). METHODS We studied 42 patients with t(8;21)(q22;q22)/RUNX1-RUNX1T1 and 51 with inv(16)(p13.1q22)/CBFB-MYH11 Tandem MRD analyses by MFC and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were performed in 281 bone marrow (BM) samples. RESULTS Grouping qRT-PCR levels as ≤0.01, 0.01 to 0.1, 0.1 to 1, 1 to 10, and >10%, and reporting MFC (sensitivity, 0.1%-0.01%) as positive or negative, κ coefficient test showed no agreement between qRT-PCR and MFC in BM samples obtained postinduction (n = 44, κ = 0.041), and only weak agreement during consolidation (n = 108, κ = 0.083), maintenance/follow-up (n = 107, κ = 0.164), and salvage chemotherapy (n = 24, 0.376). In the post induction BM samples, while qRT-PCR <0.1% was associated with lower and ≥10% with higher AML relapse risk (P = .035), qRT-PCR between 0.1% to 1% and 1% to 10% failed to predict relapse. In the latter group with intermediate qRT-PCR results, MFC provided prognostic value for relapse (P = 0.006). CONCLUSIONS MFC and qRT-PCR are complementary tests in monitoring CBF AML MRD.
Collapse
Affiliation(s)
| | | | - Jie Peng
- From the Departments of Hematopathology
| | | | - Naval Daver
- Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Gautam Borthakur
- Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | | | | | | | - Farhad Ravandi
- Leukemia, The University of Texas MD Anderson Cancer Center, Houston
| | - Sa A Wang
- From the Departments of Hematopathology
| |
Collapse
|
40
|
Yu YRA, Hotten DF, Malakhau Y, Volker E, Ghio AJ, Noble PW, Kraft M, Hollingsworth JW, Gunn MD, Tighe RM. Flow Cytometric Analysis of Myeloid Cells in Human Blood, Bronchoalveolar Lavage, and Lung Tissues. Am J Respir Cell Mol Biol 2016; 54:13-24. [PMID: 26267148 DOI: 10.1165/rcmb.2015-0146oc] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Clear identification of specific cell populations by flow cytometry is important to understand functional roles. A well-defined flow cytometry panel for myeloid cells in human bronchoalveolar lavage (BAL) and lung tissue is currently lacking. The objective of this study was to develop a flow cytometry-based panel for human BAL and lung tissue. We obtained and performed flow cytometry/sorting on human BAL cells and lung tissue. Confocal images were obtained from lung tissue using antibodies for cluster of differentiation (CD)206, CD169, and E cadherin. We defined a multicolor flow panel for human BAL and lung tissue that identifies major leukocyte populations. These include macrophage (CD206(+)) subsets and other CD206(-) leukocytes. The CD206(-) cells include: (1) three monocyte (CD14(+)) subsets, (2) CD11c(+) dendritic cells (CD14(-), CD11c(+), HLA-DR(+)), (3) plasmacytoid dendritic cells (CD14(-), CD11c(-), HLA-DR(+), CD123(+)), and (4) other granulocytes (neutrophils, mast cells, eosinophils, and basophils). Using this panel on human lung tissue, we defined two populations of pulmonary macrophages: CD169(+) and CD169(-) macrophages. In lung tissue, CD169(-) macrophages were a prominent cell type. Using confocal microscopy, CD169(+) macrophages were located in the alveolar space/airway, defining them as alveolar macrophages. In contrast, CD169(-) macrophages were associated with airway/alveolar epithelium, consistent with interstitial-associated macrophages. We defined a flow cytometry panel in human BAL and lung tissue that allows identification of multiple immune cell types and delineates alveolar from interstitial-associated macrophages. This study has important implications for defining myeloid cells in human lung samples.
Collapse
Affiliation(s)
- Yen-Rei A Yu
- 1 Department of Medicine, Duke University, Durham, North Carolina
| | | | - Yuryi Malakhau
- 1 Department of Medicine, Duke University, Durham, North Carolina
| | - Ellen Volker
- 1 Department of Medicine, Duke University, Durham, North Carolina
| | - Andrew J Ghio
- 2 National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Chapel Hill, North Carolina
| | - Paul W Noble
- 3 Department of Medicine, Cedar Sinai Medical Center, Los Angeles, California; and
| | - Monica Kraft
- 1 Department of Medicine, Duke University, Durham, North Carolina
| | | | - Michael D Gunn
- 1 Department of Medicine, Duke University, Durham, North Carolina
| | - Robert M Tighe
- 1 Department of Medicine, Duke University, Durham, North Carolina
| |
Collapse
|
41
|
Tierens A, Bjørklund E, Siitonen S, Marquart HV, Wulff-Juergensen G, Pelliniemi TT, Forestier E, Hasle H, Jahnukainen K, Lausen B, Jonsson OG, Palle J, Zeller B, Fogelstrand L, Abrahamsson J. Residual disease detected by flow cytometry is an independent predictor of survival in childhood acute myeloid leukaemia; results of the NOPHO-AML 2004 study. Br J Haematol 2016; 174:600-9. [PMID: 27072379 DOI: 10.1111/bjh.14093] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/15/2016] [Indexed: 01/10/2023]
Abstract
Early response after induction is a prognostic factor for disease outcome in childhood acute myeloid leukaemia (AML). Residual disease (RD) detection by multiparameter flow cytometry (MFC) was performed at day 15 and before consolidation therapy in 101 patients enrolled in the Nordic Society of Paediatric Haemato-Oncology AML 2004 study. A multicentre laboratory approach to RD analysis was used. Event-free survival (EFS) and overall survival (OS) was significantly different in patients with and without RD at both time points, using a 0·1% RD cut-off level. RD-negative and -positive patients after first induction showed a 5-year EFS of 65 ± 7% and 22 ± 7%, respectively (P < 0·001) and an OS of 77 ± 6% (P = 0·025) and 51 ± 8%. RD-negative and -positive patients at start of consolidation therapy had a 5-year EFS of 57 ± 7% and 11 ± 7%, respectively (P < 0·001) and an OS of 78 ± 6% and 28 ± 11%) (P < 0·001). In multivariate analysis only RD was significantly correlated with survival. RD before consolidation therapy was the strongest independent prognostic factor for EFS [hazard ratio (HR):5·0; 95% confidence interval (CI):1·9-13·3] and OS (HR:7·0; 95%CI:2·0-24·5). In conclusion, RD before consolidation therapy identifies patients at high risk of relapse in need of intensified treatment. In addition, RD detection can be performed in a multicentre setting and can be implemented in future trials.
Collapse
Affiliation(s)
- Anne Tierens
- Department of Pathobiology and Laboratory Medicine, University Health Network, Toronto General Hospital, Toronto, ON, Canada.,Department of Pathology, Oslo University Hospital, Oslo, Norway
| | | | - Sanna Siitonen
- Laboratory Services (Hospital District of Helsinki and Uusimaa Laboratory), Helsinki University Central Hospital, Helsinki, Finland
| | - Hanne Vibeke Marquart
- Department of Clinical Immunology section 7631, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Gitte Wulff-Juergensen
- Department of Clinical Immunology section 7631, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | | | - Henrik Hasle
- Aarhus University, Hospital Skejby, Aarhus, Denmark
| | - Kirsi Jahnukainen
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | | | | - Bem Zeller
- Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Linda Fogelstrand
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jonas Abrahamsson
- Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
42
|
Jackson JM, Taylor JB, Witek MA, Hunsucker SA, Waugh JP, Fedoriw Y, Shea TC, Soper SA, Armistead PM. Microfluidics for the detection of minimal residual disease in acute myeloid leukemia patients using circulating leukemic cells selected from blood. Analyst 2016; 141:640-51. [PMID: 26523411 PMCID: PMC4701594 DOI: 10.1039/c5an01836f] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We report a highly sensitive microfluidic assay to detect minimal residual disease (MRD) in patients with acute myeloid leukemia (AML) that samples peripheral blood to search for circulating leukemic cells (CLCs). Antibodies immobilized within three separate microfluidic devices affinity-selected CLC subpopulations directly from peripheral blood without requiring pre-processing. The microfluidic devices targeted CD33, CD34, and CD117 cell surface antigens commonly expressed by AML leukemic cells so that each subpopulation's CLC numbers could be tracked to determine the onset of relapse. Staining against aberrant markers (e.g. CD7, CD56) identified low levels (11-2684 mL(-1)) of CLCs. The commonly used platforms for the detection of MRD for AML patients are multi-parameter flow cytometry (MFC), typically from highly invasive bone marrow biopsies, or PCR from blood samples, which is limited to <50% of AML patients. In contrast, the microfluidic assay is a highly sensitive blood test that permits frequent sampling for >90% of all AML patients using the markers selected for this study (selection markers CD33, CD34, CD117 and aberrant markers such as CD7 and CD56). We present data from AML patients after stem cell transplant (SCT) therapy using our assay. We observed high agreement of the microfluidic assay with therapeutic treatment and overall outcome. We could detect MRD at an earlier stage compared to both MFC and PCR directly from peripheral blood, obviating the need for a painful bone marrow biopsy. Using the microfluidic assay, we detected MRD 28 days following one patient's SCT and the onset of relapse at day 57, while PCR from a bone marrow biopsy did not detect MRD until day 85 for the same patient. Earlier detection of MRD in AML post-SCT enabled by peripheral blood sampling using the microfluidic assay we report herein can influence curative clinical decisions for AML patients.
Collapse
MESH Headings
- Animals
- Hematopoietic Stem Cell Transplantation
- Humans
- Lab-On-A-Chip Devices
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/surgery
- Neoplasm, Residual/blood
- Neoplasm, Residual/diagnosis
- Neoplasm, Residual/pathology
- Neoplastic Cells, Circulating/pathology
- Recurrence
- Sensitivity and Specificity
Collapse
Affiliation(s)
- Joshua M Jackson
- Department of Chemistry, UNC-Chapel Hill, USA. and Center for Biomodular Multi-scale Systems for Precision Medicine, UNC-Chapel Hill, USA
| | - James B Taylor
- Department of Chemistry, UNC-Chapel Hill, USA. and Center for Biomodular Multi-scale Systems for Precision Medicine, UNC-Chapel Hill, USA
| | - Małgorzata A Witek
- Center for Biomodular Multi-scale Systems for Precision Medicine, UNC-Chapel Hill, USA and Department of Biomedical Engineering, UNC-Chapel Hill, USA
| | - Sally A Hunsucker
- University of North Carolina Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, USA.
| | | | - Yuri Fedoriw
- University of North Carolina Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, USA. and Department of Medicine, UNC-Chapel Hill, USA
| | | | - Steven A Soper
- Department of Chemistry, UNC-Chapel Hill, USA. and Center for Biomodular Multi-scale Systems for Precision Medicine, UNC-Chapel Hill, USA
| | - Paul M Armistead
- University of North Carolina Lineberger Comprehensive Cancer Center, UNC-Chapel Hill, USA. and Department of Medicine, UNC-Chapel Hill, USA
| |
Collapse
|
43
|
The role of multiparametric flow cytometry in the detection of minimal residual disease in acute leukaemia. Pathology 2015; 47:609-21. [DOI: 10.1097/pat.0000000000000319] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
44
|
Abstract
PURPOSE OF REVIEW Acute myeloid leukemia (AML) is a heterogeneous disease. Detection of minimal residual disease (MRD) has the potential to improve risk stratification, and its routine monitoring may allow timely therapeutic actions such as allogeneic hematopoietic stem cell transplantation. The current review will discuss challenges and available evidence for clinical application of MRD detection in AML management. RECENT FINDINGS The heterogeneous nature of AML, variations in genetic aberrations and immunophenotypes among patients and between malignant subclones coexisting within a single patient, is a challenge for the development of a reliable MRD test in AML. MRD value was demonstrated in subtypes of AML in which reliable leukemia-specific genetic marker is present (e.g., core-binding leukemia, AML positive for NPM1 mutation). Multicolor flow cytometry and quantitative PCR monitoring for Wilms tumor 1 gene transcript have also been shown to correlate with disease progression. MRD results should always be interpreted within patient-specific clinical context considering other risk factors and timing of MRD eradication. SUMMARY Introduction of MRD testing into routine clinical practice is a challenge in AML. An improvement in laboratory techniques along with identification of additional leukemia-specific markers is required.
Collapse
|
45
|
Furman D, Davis MM. New approaches to understanding the immune response to vaccination and infection. Vaccine 2015; 33:5271-81. [PMID: 26232539 DOI: 10.1016/j.vaccine.2015.06.117] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/26/2015] [Accepted: 06/29/2015] [Indexed: 02/06/2023]
Abstract
The immune system is a network of specialized cell types and tissues that communicates via cytokines and direct contact, to orchestrate specific types of defensive responses. Until recently, we could only study immune responses in a piecemeal, highly focused fashion, on major components like antibodies to the pathogen. But recent advances in technology and in our understanding of the many components of the system, innate and adaptive, have made possible a broader approach, where both the multiple responding cells and cytokines in the blood are measured. This systems immunology approach to a vaccine response or an infection gives us a more holistic picture of the different parts of the immune system that are mobilized and should allow us a much better understanding of the pathways and mechanisms of such responses, as well as to predict vaccine efficacy in different populations well in advance of efficacy studies. Here we summarize the different technologies and methods and discuss how they can inform us about the differences between diseases and vaccines, and how they can greatly accelerate vaccine development.
Collapse
Affiliation(s)
- David Furman
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States; Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States; Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States; Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA, United States.
| |
Collapse
|
46
|
Cabral Filho PE, Pereira MIA, Fernandes HP, de Thomaz AA, Cesar CL, Santos BS, Barjas-Castro ML, Fontes A. Blood group antigen studies using CdTe quantum dots and flow cytometry. Int J Nanomedicine 2015; 10:4393-404. [PMID: 26185442 PMCID: PMC4501227 DOI: 10.2147/ijn.s84551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
New methods of analysis involving semiconductor nanocrystals (quantum dots [QDs]) as fluorescent probes have been highlighted in life science. QDs present some advantages when compared to organic dyes, such as size-tunable emission spectra, broad absorption bands, and principally exceptional resistance to photobleaching. Methods applying QDs can be simple, not laborious, and can present high sensibility, allowing biomolecule identification and quantification with high specificity. In this context, the aim of this work was to apply dual-color CdTe QDs to quantify red blood cell (RBC) antigen expression on cell surface by flow cytometric analysis. QDs were conjugated to anti-A or anti-B monoclonal antibodies, as well as to the anti-H (Ulex europaeus I) lectin, to investigate RBCs of A1, B, A1B, O, A2, and Aweak donors. Bioconjugates were capable of distinguishing the different expressions of RBC antigens, both by labeling efficiency and by flow cytometry histogram profile. Furthermore, results showed that RBCs from Aweak donors present fewer amounts of A antigens and higher amounts of H, when compared to A1 RBCs. In the A group, the amount of A antigens decreased as A1 > A3 > AX = Ael, while H antigens were AX = Ael > A1. Bioconjugates presented stability and remained active for at least 6 months. In conclusion, this methodology with high sensibility and specificity can be applied to study a variety of RBC antigens, and, as a quantitative tool, can help in achieving a better comprehension of the antigen expression patterns on RBC membranes.
Collapse
Affiliation(s)
- Paulo E Cabral Filho
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Maria I A Pereira
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Heloise P Fernandes
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Andre A de Thomaz
- Departamento de Eletrônica Quântica, Instituto de Física Gleb Wataghin, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Carlos L Cesar
- Departamento de Eletrônica Quântica, Instituto de Física Gleb Wataghin, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Beate S Santos
- Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Maria L Barjas-Castro
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Adriana Fontes
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
47
|
Ouyang J, Goswami M, Tang G, Peng J, Ravandi F, Daver N, Routbort M, Konoplev S, Lin P, Medeiros LJ, Jorgensen JL, Wang SA. The clinical significance of negative flow cytometry immunophenotypic results in a morphologically scored positive bone marrow in patients following treatment for acute myeloid leukemia. Am J Hematol 2015; 90:504-10. [PMID: 25732229 DOI: 10.1002/ajh.23988] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 02/11/2015] [Accepted: 02/18/2015] [Indexed: 12/16/2022]
Abstract
In a patient with acute myeloid leukemia (AML) following therapy, finding ≥5% bone marrow (BM) blasts is highly concerning for residual/relapsed disease. Over an 18-month period, we performed multicolor flow cytometry immunophenotyping (MFC) for AML minimal residual disease on >4,000 BM samples, and identified 41 patients who had ≥5% myeloblasts by morphology but negative by MFC. At the time of a negative MFC study, an abnormal cytogenetic study converted to negative in 14 patients and remained positive at a low level (2.5-9.5%) by fluorescence in situ hybridization in 3 (14%), of the latter, abnormalities subsequently disappeared in the repeated BM in 2 patients. Positive pretreatment mutations, including FLT3, NPM1, IDH1, CEBPA, became negative in all 10 patients tested. Of the seven patients with favorable cytogenetics, PML/RARA, CBFB-MYH11 or RUNX1-RUNX1T1 fusion transcripts were detected at various levels in six patients but all patients remained in complete remission. With no additional chemotherapy given, 39 patients had BM repeated (median 2 weeks, range <1-21), and all cases showed <5% BM blasts and a continuously negative MFC. In the end of follow-up (median 10 months, range 1-22), 13 patients experienced relapse, 12/13 showing clonal cytogenetic evolution/switch and 11 demonstrating major immunophenotypic shifts. We conclude that MFC is useful in identifying a regenerating BM sample with ≥5% BM blasts that would otherwise be scored as positive using standard morphologic examination. We believe this conclusion is supported by the changes in molecular cytogenetic status and the patient clinical follow-up data.
Collapse
Affiliation(s)
- Juan Ouyang
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
- Department of Laboratory Medicine; the First Affiliated Hospital of Sun Yat-Sen University; Guangzhou Guangdong China
| | - Maitrayee Goswami
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - Guilin Tang
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - Jie Peng
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
- Department of Hematology Oncology; the First Affiliated Hospital of Xiangya Medical School Central South University; Changsha Hunan China
| | - Farhad Ravandi
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - Naval Daver
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - Mark Routbort
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - Sergej Konoplev
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - Pei Lin
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - L. Jeffrey Medeiros
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - Jeffrey L. Jorgensen
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| | - Sa A. Wang
- Department of Hematopathology; the University of Texas MD Anderson Cancer Center; Houston Texas
| |
Collapse
|
48
|
Xu Y, Plazyo O, Romero R, Hassan SS, Gomez-Lopez N. Isolation of Leukocytes from the Human Maternal-fetal Interface. J Vis Exp 2015:e52863. [PMID: 26067211 DOI: 10.3791/52863] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pregnancy is characterized by the infiltration of leukocytes in the reproductive tissues and at the maternal-fetal interface (decidua basalis and decidua parietalis). This interface is the anatomical site of contact between maternal and fetal tissues; therefore, it is an immunological site of action during pregnancy. Infiltrating leukocytes at the maternal-fetal interface play a central role in implantation, pregnancy maintenance, and timing of delivery. Therefore, phenotypic and functional characterizations of these leukocytes will provide insight into the mechanisms that lead to pregnancy disorders. Several protocols have been described in order to isolate infiltrating leukocytes from the decidua basalis and decidua parietalis; however, the lack of consistency in the reagents, enzymes, and times of incubation makes it difficult to compare these results. Described herein is a novel approach that combines the use of gentle mechanical and enzymatic dissociation techniques to preserve the viability and integrity of extracellular and intracellular markers in leukocytes isolated from the human tissues at the maternal-fetal interface. Aside from immunophenotyping, cell culture, and cell sorting, the future applications of this protocol are numerous and varied. Following this protocol, the isolated leukocytes can be used to determine DNA methylation, expression of target genes, in vitro leukocyte functionality (i.e., phagocytosis, cytotoxicity, T-cell proliferation, and plasticity, etc.), and the production of reactive oxygen species at the maternal-fetal interface. Additionally, using the described protocol, this laboratory has been able to describe new and rare leukocytes at the maternal-fetal interface.
Collapse
Affiliation(s)
- Yi Xu
- Perinatology Research Branch, NICHD/NIH/DHHS
| | | | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS; Department of Obstetrics and Gynecology, University of Michigan; Department of Epidemiology and Biostatistics, Michigan State University; Department of Molecular Obstetrics and Genetics, Wayne State University
| | - Sonia S Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS; Department of Obstetrics and Gynecology, Wayne State University School of Medicine
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, NICHD/NIH/DHHS; Department of Obstetrics and Gynecology, Wayne State University School of Medicine; Department of Immunology and Microbiology, Wayne State University School of Medicine;
| |
Collapse
|
49
|
Goswami M, McGowan KS, Lu K, Jain N, Candia J, Hensel NF, Tang J, Calvo KR, Battiwalla M, Barrett AJ, Hourigan CS. A multigene array for measurable residual disease detection in AML patients undergoing SCT. Bone Marrow Transplant 2015; 50:642-51. [PMID: 25665046 PMCID: PMC4424111 DOI: 10.1038/bmt.2014.326] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 12/15/2014] [Accepted: 12/19/2014] [Indexed: 02/04/2023]
Abstract
AML is a diagnosis encompassing a diverse group of myeloid malignancies. Heterogeneous genetic etiology, together with the potential for oligoclonality within the individual patient, have made the identification of a single high-sensitivity marker of disease burden challenging. We developed a multiple gene measurable residual disease (MG-MRD) RQ-PCR array for the high-sensitivity detection of AML, retrospectively tested on 74 patients who underwent allo-SCT at the NHLBI in the period 1994-2012. MG-MRD testing on peripheral blood samples prior to transplantation demonstrated excellent concordance with traditional BM-based evaluation and improved risk stratification for post-transplant relapse and OS outcomes. Pre-SCT assessment by MG-MRD predicted all clinical relapses occurring in the first 100 days after allo-SCT compared with 57% sensitivity using WT1 RQ-PCR alone. Nine patients who were negative for WT1 prior to transplantation were correctly reclassified into a high-risk MG-MRD-positive group, associated with 100% post-transplant mortality. This study provides proof of principle that a multiple gene approach may be superior to the use of WT1 expression alone for AML residual disease detection.
Collapse
Affiliation(s)
- M Goswami
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - K S McGowan
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - K Lu
- Stem Cell Allogenic Transplantation Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - N Jain
- Stem Cell Allogenic Transplantation Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - J Candia
- Department of Physics, University of Maryland, College Park, MD, USA
- School of Medicine, University of Maryland, Baltimore, MD, USA
| | - N F Hensel
- Stem Cell Allogenic Transplantation Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - J Tang
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - K R Calvo
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - M Battiwalla
- Stem Cell Allogenic Transplantation Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - A J Barrett
- Stem Cell Allogenic Transplantation Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - C S Hourigan
- Myeloid Malignancies Section, Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|