CC BY 4.0 · Indian J Med Paediatr Oncol 2023; 44(05): 494-504
DOI: 10.1055/s-0043-1766137
Review Article

Flow Cytometric MRD Assessment in Acute Lymphoblastic Leukemias

Harpreet Virk
1   Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
,
Man Updesh Singh Sachdeva
1   Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
› Author Affiliations

Abstract

Acute lymphoblastic leukemia (ALL) is one of the very first malignancy where the assessment of early response to therapy by minimal/measurable residual disease (MRD) monitoring has proven to be cardinal tool for guiding therapeutic choices. At present, MRD detection is not only used for the assessment of initial treatment response and subsequent risk stratification but also for monitoring disease burden in the setting of hematopoietic stem cell transplant. Multicolor flow cytometry (FCM) for the assessment of MRD has been in existence for more than two decades. It is presently the most commonly used technique worldwide for MRD assessment in ALL. The technique has evolved from two to three color assays in its early phases to eight and more color assays in present time, which enables detection of one leukemic cell in 104 or more cells. The assessment of MRD is based on analysis of expression of lineage-associated markers and either looking at “leukemia associated immunophenotypes” or identify “different from normal” patterns. A rapid turn-around-time and direct quantification of viable residual leukemic cells are advantages of FCM over molecular techniques of MRD assessment. On the other hand, one of the prime limitations of detection of residual cells by FCM is the immunophenotypic shifts that are observed as a result of chemotherapeutic reagents. In addition, introduction of immunotherapy, especially against important gating markers like CD19, has posed significant challenge to FCM-based MRD assays, and requires modification of antibody panels for an alternate gating and analysis strategy. Finally, standardization and validation of MRD assay and use of internal and external quality controls are extremely important aspects for a clinical laboratory providing MRD reports for patient care.



Publication History

Article published online:
17 April 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

 
  • References

  • 1 Terwilliger T, Abdul-Hay M. Acute lymphoblastic leukemia: a comprehensive review and 2017 update. Blood Cancer J 2017; 7 (06) e577
  • 2 Rafei H, Kantarjian HM, Jabbour EJ. Recent advances in the treatment of acute lymphoblastic leukemia. Leuk Lymphoma 2019; 60 (11) 2606-2621
  • 3 Jabbour E, O'Brien S, Konopleva M, Kantarjian H. New insights into the pathophysiology and therapy of adult acute lymphoblastic leukemia. Cancer 2015; 121 (15) 2517-2528
  • 4 Swerdlow SH, Campo E, Harris NL. et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Vol. Revised 4th Edition. Lyon: WHO; 2017
  • 5 Shafique S, Tehsin S. Acute lymphoblastic leukemia detection and classification of its subtypes using pretrained deep convolutional neural networks. Technol Cancer Res Treat 2018; 17: 1533033818802789
  • 6 Einsiedel HG, Von Stackelberg A, Hartmann R. et al. Long-term outcome in NGS for MRD assessment in ALL 489 children with relapsed ALL by risk-stratified salvage therapy: results of trial acute lymphoblastic leukemia-relapse study of the Berlin-Frankfurt-Münster Group 87. J Clin Oncol 2005; 23: 7942-7950
  • 7 Gökbuget N, Stanze D, Beck J. et al; German Multicenter Study Group for Adult Acute Lymphoblastic Leukemia. Outcome of relapsed adult lymphoblastic leukemia depends on response to salvage chemotherapy, prognostic factors, and performance of stem cell transplantation. Blood 2012; 120 (10) 2032-2041
  • 8 Inaba H, Coustan-Smith E, Cao X. et al. Comparative analysis of different approaches to measure treatment response in acute myeloid leukemia. J Clin Oncol 2012; 30 (29) 3625-3632
  • 9 Loken MR, Alonzo TA, Pardo L. et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: a report from Children's Oncology Group. Blood 2012; 120 (08) 1581-1588
  • 10 Freeman SD, Virgo P, Couzens S. et al. Prognostic relevance of treatment response measured by flow cytometric residual disease detection in older patients with acute myeloid leukemia. J Clin Oncol 2013; 31 (32) 4123-4131
  • 11 van der Velden VH, Hochhaus A, Cazzaniga G, Szczepanski T, Gabert J, van Dongen JJ. Detection of minimal residual disease in hematologic malignancies by real-time quantitative PCR: principles, approaches, and laboratory aspects. Leukemia 2003; 17 (06) 1013-1034
  • 12 van der Velden VH, Cazzaniga G, Schrauder A. et al; European Study Group on MRD detection in ALL (ESG-MRD-ALL). Analysis of minimal residual disease by Ig/TCR gene rearrangements: guidelines for interpretation of real-time quantitative PCR data. Leukemia 2007; 21 (04) 604-611
  • 13 Campana D. Status of minimal residual disease testing in childhood haematological malignancies. Br J Haematol 2008; 143 (04) 481-489
  • 14 Flohr T, Schrauder A, Cazzaniga G. et al; International BFM Study Group (I-BFM-SG). Minimal residual disease-directed risk stratification using real-time quantitative PCR analysis of immunoglobulin and T-cell receptor gene rearrangements in the international multicenter trial AIEOP-BFM ALL 2000 for childhood acute lymphoblastic leukemia. Leukemia 2008; 22 (04) 771-782
  • 15 Dworzak MN, Gaipa G, Ratei R. et al. Standardization of flow cytometric minimal residual disease evaluation in acute lymphoblastic leukemia: multicentric assessment is feasible. Cytometry B Clin Cytom 2008; 74 (06) 331-340
  • 16 Chen X, Wood BL. Monitoring minimal residual disease in acute leukemia: technical challenges and interpretive complexities. Blood Rev 2017; 31 (02) 63-75
  • 17 Fronkova E, Muzikova K, Mejstrikova E. et al. B-cell reconstitution after allogeneic SCT impairs minimal residual disease monitoring in children with ALL. Bone Marrow Transplant 2008; 42 (03) 187-196
  • 18 Dworzak MN, Gaipa G, Schumich A. et al. Modulation of antigen expression in B-cell precursor acute lymphoblastic leukemia during induction therapy is partly transient: evidence for a drug-induced regulatory phenomenon. Results of the AIEOP-BFM-ALL-FLOW-MRD-Study Group. Cytometry B Clin Cytom 2010; 78 (03) 147-153
  • 19 Fronkova E, Mejstrikova E, Avigad S. et al. Minimal residual disease (MRD) analysis in the non-MRD-based ALL IC-BFM 2002 protocol for childhood ALL: is it possible to avoid MRD testing?. Leukemia 2008; 22 (05) 989-997
  • 20 Borowitz MJ, Devidas M, Hunger SP. et al; Children's Oncology Group. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia and its relationship to other prognostic factors: a Children's Oncology Group study. Blood 2008; 111 (12) 5477-5485
  • 21 Basso G, Veltroni M, Valsecchi MG. et al. Risk of relapse of childhood acute lymphoblastic leukemia is predicted by flow cytometric measurement of residual disease on day 15 bone marrow. J Clin Oncol 2009; 27 (31) 5168-5174
  • 22 Sutton R, Venn NC, Tolisano J. et al; Australian and New Zealand Children's Oncology Group. Clinical significance of minimal residual disease at day 15 and at the end of therapy in childhood acute lymphoblastic leukaemia. Br J Haematol 2009; 146 (03) 292-299
  • 23 Coustan-Smith E, Sancho J, Hancock ML. et al. Use of peripheral blood instead of bone marrow to monitor residual disease in children with acute lymphoblastic leukemia. Blood 2002; 100 (07) 2399-2402
  • 24 Pui CH, Mullighan CG, Evans WE, Relling MV. Pediatric acute lymphoblastic leukemia: where are we going and how do we get there?. Blood 2012; 120 (06) 1165-1174
  • 25 Berry DA, Zhou S, Higley H. et al. Association of minimal residual disease with clinical outcome in pediatric and adult acute lympho-blastic leukemia: a meta-analysis. JAMA Oncol 2017; 3 (07) e170580
  • 26 Jeha S, Pui CH. Risk-adapted treatment of pediatric acute lymphoblastic leukemia. Hematol Oncol Clin North Am 2009; 23 (05) 973-990 , v
  • 27 Bommannan K, Singh Sachdeva MU, Varma N, Bose P, Bansal D. Role of Mid-induction peripheral blood minimal residual disease detection in pediatric B-lineage acute lymphoblastic leukemia. Indian Pediatr 2016; 53 (12) 1065-1068
  • 28 Cavé H, van der Werff ten Bosch J, Suciu S. et al; European Organization for Research and Treatment of Cancer-Childhood Leukemia Cooperative Group. Clinical significance of minimal residual disease in childhood acute lymphoblastic leukemia. European Organization for Research and Treatment of Cancer–Childhood Leukemia Cooperative Group. N Engl J Med 1998; 339 (09) 591-598
  • 29 Kotrova M, Volland A, Kehden B. et al. Comparison of minimal residual disease levels in bone marrow and peripheral blood in adult acute lymphoblastic leukemia. Leukemia 2020; 34 (04) 1154-1157
  • 30 Helgestad J, Rosthøj S, Johansen P, Varming K, Østergaard E. Bone marrow aspiration technique may have an impact on therapy stratification in children with acute lymphoblastic leukaemia. Pediatr Blood Cancer 2011; 57 (02) 224-226
  • 31 Theunissen P, Mejstrikova E, Sedek L. et al; EuroFlow Consortium. Standardized flow cytometry for highly sensitive MRD measurements in B-cell acute lymphoblastic leukemia. Blood 2017; 129 (03) 347-357
  • 32 Chatterjee G, Sriram H, Ghogale S. et al. Mimics and artefacts of measurable residual disease in a highly sensitive multicolour flow cytometry assay for B-lymphoblastic leukaemia/lymphoma: critical consideration for analysis of measurable residual disease. Br J Haematol 2022; 196 (02) 374-379
  • 33 Wood BL. Principles of minimal residual disease detection for hematopoietic neoplasms by flow cytometry. Cytometry B Clin Cytom 2016; 90 (01) 47-53
  • 34 van Dongen JJ, Seriu T, Panzer-Grümayer ER. et al. Prognostic value of minimal residual disease in acute lymphoblastic leukaemia in childhood. Lancet 1998; 352 (9142): 1731-1738
  • 35 Zhou J, Goldwasser MA, Li A. et al; Dana-Farber Cancer Institute ALL Consortium. Quantitative analysis of minimal residual disease predicts relapse in children with B-lineage acute lymphoblastic leukemia in DFCI ALL Consortium Protocol 95-01. Blood 2007; 110 (05) 1607-1611
  • 36 Vora A, Goulden N, Mitchell C. et al. Augmented post-remission therapy for a minimal residual disease-defined high-risk subgroup of children and young people with clinical standard-risk and intermediate-risk acute lymphoblastic leukaemia (UKALL 2003): a randomised controlled trial. Lancet Oncol 2014; 15 (08) 809-818
  • 37 Das N, Banavali S, Bakhshi S. et al. Protocol for ICiCLe-ALL-14 (InPOG-ALL-15-01): a prospective, risk stratified, randomised, multicentre, open label, controlled therapeutic trial for newly diagnosed childhood acute lymphoblastic leukaemia in India. Trials 2022; 23 (01) 102
  • 38 Lúcio P, Parreira A, van den Beemd MW. et al. Flow cytometric analysis of normal B cell differentiation: a frame of reference for the detection of minimal residual disease in precursor-B-ALL. Leukemia 1999; 13 (03) 419-427
  • 39 Ciudad J, San Miguel JF, López-Berges MC. et al. Detection of abnormalities in B-cell differentiation pattern is a useful tool to predict relapse in precursor-B-ALL. Br J Haematol 1999; 104 (04) 695-705
  • 40 Denys B, van der Sluijs-Gelling AJ, Homburg C. et al. Improved flow cytometric detection of minimal residual disease in childhood acute lymphoblastic leukemia. Leukemia 2013; 27 (03) 635-641
  • 41 Karawajew L, Dworzak M, Ratei R. et al. Minimal residual disease analysis by eight-color flow cytometry in relapsed childhood acute lymphoblastic leukemia. Haematologica 2015; 100 (07) 935-944
  • 42 Bouriche L, Bernot D, Nivaggioni V, Arnoux I, Loosveld M. Detection of minimal residual disease in B cell acute lymphoblastic leukemia using an eight-color tube with dried antibody reagents. Cytometry B Clin Cytom 2019; 96 (02) 158-163
  • 43 Nováková M, Glier H, Brdičková N. et al. How to make usage of the standardized EuroFlow 8-color protocols possible for instruments of different manufacturers. J Immunol Methods 2019; 475: 112388
  • 44 Solly F, Angelot F, Garand R. et al. CD304 is preferentially expressed on a subset of B-lineage acute lymphoblastic leukemia and represents a novel marker for minimal residual disease detection by flow cytometry. Cytometry A 2012; 81 (01) 17-24
  • 45 Wang W, Gao L, Li Y. et al. The application of CD73 in minimal residual disease monitoring using flow cytometry in B-cell acute lymphoblastic leukemia. Leuk Lymphoma 2016; 57 (05) 1174-1181
  • 46 Kalina T, Vaskova M, Mejstrikova E. et al. Myeloid antigens in childhood lymphoblastic leukemia: clinical data point to regulation of CD66c distinct from other myeloid antigens. BMC Cancer 2005; 5: 38
  • 47 Djokic M, Björklund E, Blennow E, Mazur J, Söderhäll S, Porwit A. Overexpression of CD123 correlates with the hyperdiploid genotype in acute lymphoblastic leukemia. Haematologica 2009; 94 (07) 1016-1019
  • 48 Behm FG, Smith FO, Raimondi SC, Pui CH, Bernstein ID. Human homologue of the rat chondroitin sulfate proteoglycan, NG2, detected by monoclonal antibody 7.1, identifies childhood acute lymphoblastic leukemias with t(4;11)(q21;q23) or t(11;19)(q23;p13) and MLL gene rearrangements. Blood 1996; 87 (03) 1134-1139
  • 49 Coustan-Smith E, Song G, Clark C. et al. New markers for minimal residual disease detection in acute lymphoblastic leukemia. Blood 2011; 117 (23) 6267-6276
  • 50 Sędek Ł, Bulsa J, Sonsala A. et al. The immunophenotypes of blast cells in B-cell precursor acute lymphoblastic leukemia: how different are they from their normal counterparts?. Cytometry B Clin Cytom 2014; 86 (05) 329-339
  • 51 McKenna RW, Washington LT, Aquino DB, Picker LJ, Kroft SH. Immunophenotypic analysis of hematogones (B-lymphocyte precursors) in 662 consecutive bone marrow specimens by 4-color flow cytometry. Blood 2001; 98 (08) 2498-2507
  • 52 Borowitz MJ, Shuster JJ, Civin CI. et al. Prognostic significance of CD34 expression in childhood B-precursor acute lymphocytic leukemia: a Pediatric Oncology Group study. J Clin Oncol 1990; 8 (08) 1389-1398
  • 53 Ali Shah M, Ahmad U, Tariq Mahmood M, Ahmad AH, Abu Bakar M. Frequency of CD34 and CD10 expression in adolescent and young adult patients having precursor B-cell acute lymphoblastic leukemia and its correlation with clinical outcomes: a single-center study. Cureus 2022; 14 (01) e21261
  • 54 Garg N, Gupta R, Kotru M. CD34 is not expressed by blasts in a third of B-ALL patients and its negativity is associated with aberrant marker expression: a retrospective analysis. Asian Pac J Cancer Prev 2021; 22 (03) 919-925
  • 55 Gaipa G, Basso G, Aliprandi S. et al; I-BFM-ALL-FCM-MRD-Study Group. Prednisone induces immunophenotypic modulation of CD10 and CD34 in nonapoptotic B-cell precursor acute lymphoblastic leukemia cells. Cytometry B Clin Cytom 2008; 74 (03) 150-155
  • 56 Chantepie SP, Cornet E, Salaün V, Reman O. Hematogones: an overview. Leuk Res 2013; 37 (11) 1404-1411
  • 57 Shaver AC, Seegmiller AC. Lymphoblastic leukemia minimal residual disease assessment by flow cytometric analysis. Clin Lab Med 2017; 37 (04) 771-785
  • 58 Sędek Ł, Theunissen P, Sobral da Costa E. et al; EuroFlow Consortium. Differential expression of CD73, CD86 and CD304 in normal vs. leukemic B-cell precursors and their utility as stable minimal residual disease markers in childhood B-cell precursor acute lymphoblastic leukemia. J Immunol Methods 2019; 475: 112429
  • 59 Chatterjee G, Sriram H, Ghogale S. et al. Immunophenotypic shift in the B-cell precursors from regenerating bone marrow samples: a critical consideration for measurable residual disease assessment in B-lymphoblastic leukemia. Cytometry B Clin Cytom 2021; 100 (04) 434-445
  • 60 Topp MS, Gökbuget N, Stein AS. et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol 2015; 16 (01) 57-66
  • 61 Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 2015; 125 (26) 4017-4023
  • 62 Cherian S, Miller V, McCullouch V, Dougherty K, Fromm JR, Wood BL. A novel flow cytometric assay for detection of residual disease in patients with B-lymphoblastic leukemia/lymphoma post anti-CD19 therapy. Cytometry B Clin Cytom 2018; 94 (01) 112-120
  • 63 O'Brien MM, Ji L, Shah NN. et al. Phase II trial of inotuzumab ozogamicin in children and adolescents with relapsed or refractory B-cell acute lymphoblastic leukemia: Children's Oncology Group Protocol AALL1621. J Clin Oncol 2022; 40 (09) 956-967
  • 64 Mikhailova E, Gluhanyuk E, Illarionova O. et al. Immunophenotypic changes of leukemic blasts in children with relapsed/refractory B-cell precursor acute lymphoblastic leukemia, who have been treated with Blinatumomab. Haematologica 2021; 106 (07) 2009-2012
  • 65 Demaret J, Varlet P, Trauet J. et al. Monitoring CAR T-cells using flow cytometry. Cytometry B Clin Cytom 2021; 100 (02) 218-224
  • 66 Brammer JE, Saliba RM, Jorgensen JL. et al. Multi-center analysis of the effect of T-cell acute lymphoblastic leukemia subtype and minimal residual disease on allogeneic stem cell transplantation outcomes. Bone Marrow Transplant 2017; 52 (01) 20-27
  • 67 Wood BL, Dunsmore KP, Devidas M. et al. T-lymphoblastic leukemia (T-ALL) shows excellent outcome, lack of significance of the early thymic precursor (ETP) immunophenotype, and validation of the prognostic value of end-induction minimal residual disease (MRD) in Children's Oncology Group (COG) study AALL0434. Blood 2014; 124: 1
  • 68 Yeoh AE, Ariffin H, Chai EL. et al. Minimal residual disease-guided treatment deintensification for children with acute lymphoblastic leukemia: results from the Malaysia-Singapore acute lymphoblastic leukemia 2003 study. J Clin Oncol 2012; 30 (19) 2384-2392
  • 69 Wang K, Chen X, Wuxiao Z. et al. Long-term outcomes of modified Berlin-Frankfurt-Münster-90 regimen in adults with T-lymphoblastic lymphoma: a single-center experience. Leuk Lymphoma 2014; 55 (08) 1800-1805
  • 70 Modvig S, Madsen HO, Siitonen SM. et al. Minimal residual disease quantification by flow cytometry provides reliable risk stratification in T-cell acute lymphoblastic leukemia. Leukemia 2019; 33 (06) 1324-1336
  • 71 van der Velden VH, Jacobs DC, Wijkhuijs AJ. et al. Minimal residual disease levels in bone marrow and peripheral blood are comparable in children with T cell acute lymphoblastic leukemia (ALL), but not in precursor-B-ALL. Leukemia 2002; 16 (08) 1432-1436
  • 72 Tembhare PR, Chatterjee G, Khanka T. et al. Eleven-marker 10-color flow cytometric assessment of measurable residual disease for T-cell acute lymphoblastic leukemia using an approach of exclusion. Cytometry B Clin Cytom 2021; 100 (04) 421-433
  • 73 Porwit-MacDonald A, Björklund E, Lucio P. et al. BIOMED-1 concerted action report: flow cytometric characterization of CD7+ cell subsets in normal bone marrow as a basis for the diagnosis and follow-up of T cell acute lymphoblastic leukemia (T-ALL). Leukemia 2000; 14 (05) 816-825
  • 74 Roshal M, Fromm JR, Winter S, Dunsmore K, Wood BL. Immaturity associated antigens are lost during induction for T cell lymphoblastic leukemia: implications for minimal residual disease detection. Cytometry B Clin Cytom 2010; 78 (03) 139-146
  • 75 DiGiuseppe JA, Wood BL. Applications of flow cytometric immunophenotyping in the diagnosis and posttreatment monitoring of B and T lymphoblastic leukemia/lymphoma. Cytometry B Clin Cytom 2019; 96 (04) 256-265
  • 76 Wood BL, Levin G, Wilson M. et al. High-throughput screening by flow cytometry identifies reduced expression of CD48 as a universal characteristic of T-ALL and a suitable target for minimal residual disease (MRD) detection. Blood 2011; 118: 2547
  • 77 Wood B, Jevremovic D, Béné MC, Yan M, Jacobs P, Litwin V. ICSH/ICCS Working Group. Validation of cell-based fluorescence assays: practice guidelines from the ICSH and ICCS - part V - assay performance criteria. Cytometry B Clin Cytom 2013; 84 (05) 315-323
  • 78 Illingworth A, Marinov I, Sutherland DR, Wagner-Ballon O, DelVecchio L. ICCS/ESCCA consensus guidelines to detect GPI-deficient cells in paroxysmal nocturnal hemoglobinuria (PNH) and related disorders part 3 - data analysis, reporting and case studies. Cytometry B Clin Cytom 2018; 94 (01) 49-66
  • 79 van Wering ER, van der Linden-Schrever BE, Szczepański T. et al. Regenerating normal B-cell precursors during and after treatment of acute lymphoblastic leukaemia: implications for monitoring of minimal residual disease. Br J Haematol 2000; 110 (01) 139-146
  • 80 Theunissen PMJ, van den Branden A, Van Der Sluijs-Gelling A. et al. Understanding the reconstitution of the B-cell compartment in bone marrow and blood after treatment for B-cell precursor acute lymphoblastic leukaemia. Br J Haematol 2017; 178 (02) 267-278
  • 81 Burnusuzov HA, Spasova MI, Murdjeva MA. et al. Immunophenotypic modulation of the blast cells in childhood acute lymphoblastic leukemia minimal residual disease detection. Folia Med (Plovdiv) 2016; 58 (01) 28-35
  • 82 Tembhare P, Badrinath Y, Ghogale S, Subramanian PG. Method for DNA ploidy analysis along with immunophenotyping for rare populations in a sample using FxCycle Violet. Curr Protoc Cytom 2017; 80: 38.1 , 15
  • 83 Preffer F, Dombkowski D. Advances in complex multiparameter flow cytometry technology: Applications in stem cell research. Cytometry B Clin Cytom 2009; 76 (05) 295-314
  • 84 Reyes-Barron C, Burack WR, Rothberg PG, Ding Y. Next-generation sequencing for minimal residual disease surveillance in acute lymphoblastic leukemia: an update. Crit Rev Oncog 2017; 22 (5-6): 559-567
  • 85 Della Starza I, Chiaretti S, De Propris MS. et al. Minimal residual disease in acute lymphoblastic leukemia: technical and clinical advances. Front Oncol 2019; 9: 726
  • 86 Yang F, Anekpuritanang T, Press RD. Clinical utility of next-generation sequencing in acute myeloid leukemia. Mol Diagn Ther 2020; 24 (01) 1-13
  • 87 Voso MT, Ottone T, Lavorgna S. et al. MRD in AML: the role of new techniques. Front Oncol 2019; 9: 655