Skip to main content
Leukemia Supplements logoLink to Leukemia Supplements
. 2012 Aug 9;1(Suppl 2):S3–S4. doi: 10.1038/leusup.2012.5

Minimal residual disease

D Campana 1,*
PMCID: PMC4851210  PMID: 27175241

Abstract

The aim of minimal residual disease (MRD) studies in patients with acute leukemia is to measure initial treatment response accurately, provide an assessment of the residual leukemic burden throughout therapy and detect relapse early. Therefore, information resulting from MRD monitoring can substantially improve many facets of clinical management. Methods for MRD detection, namely flow cytometry and PCR, have been applied to study the remission status of thousands of patients with acute lymphoblastic leukemia and acute myeloid leukemia. Collectively, the data indicate that MRD is a powerful prognostic indicator and an indispensable parameter for risk-adapted therapy. The current status of MRD in acute leukemia is briefly reviewed in this paper.

Keywords: acute lymphoblastic leukemia, acute myeloid leukemia, minimal residual disease, flow cytometry, PCR


Tracking residual leukemic cells beyond the resolution of morphological methods, that is, ‘minimal residual disease' (MRD), provides powerful prognostic information. Therefore, MRD is increasingly used to guide the clinical management of patients with acute lymphoblastic leukemia (ALL) or acute myeloid leukemia (AML).

Leukemic cells have unique genetic and immunophenotypic signatures, which can be detected by using real-time PCR or flow cytometry. In ALL, the main targets for PCR are clonal antigen-receptor (immunoglobulin and/or T-cell receptor) gene rearrangements and gene fusion transcripts (for example, BCR-ABL1, MLL-AFF1, TCF3-PBX1 and ETV6-RUNX1), which are present in about 90% and 40% of cases, respectively; they allow a sensitivity of detection of 0.001%.1 In AML, antigen-receptor gene rearrangements are normally in a germline configuration and hence cannot be used for MRD studies. The fusion transcripts most extensively used to monitor MRD in AML (in addition to PML-RARA for acute promyelocytic leukemia (APL)) are AML1-ETO, CBFbeta-MYH11 and MLL-AF9, which are present in approximately one-third of non-APL AML cases.2 In most cases of ALL and AML, cells also express marker combinations that are sufficiently distinct to allow MRD studies by flow cytometry. In the St Jude Total XV study for pediatric ALL, we could monitor MRD by flow cytometry with a 0.01% sensitivity in 482 of the 492 patients (98%),3 whereas in the St Jude AML02 trial for pediatric AML, we found aberrant immunophenotypes that allow a sensitivity of at least 0.1% in 200 of the 210 (95%) patients.4

MRD tests provide unique and important prognostic information in patients with newly diagnosed ALL, which often abolishes the prognostic weight of presenting clinical and biologic features.3, 5, 6, 7, 8 MRD is also prognostic in subgroups of ALL patients, such as infants,9 those with T-ALL10 and those with specific genetic abnormalities.7 Poorer early clearance of ALL reflected by higher MRD levels during remission induction therapy is generally associated with a higher risk of relapse.6, 11, 12 Even very low levels of MRD (that is, <0.01%) at the end of remission-induction therapy are associated with an increased risk of relapse,13 whereas high levels (that is, ⩾1%) predict a particularly poor outcome.3, 5 Moreover, MRD is a strong prognostic factor for children and adolescents with first-relapse ALL who achieve a second remission14 and for those undergoing allogeneic hematopoietic stem cell transplantation.15, 16 There is also evidence that MRD is clinically important in adult ALL. In studies of the GMALL, MRD on days 11 and 24 of treatments predicted outcome in adult patients with ‘standard-risk' ALL,17 and sequential studies of MRD were also informative.18 Other studies also demonstrated the predictive value of MRD in different subgroups of adult ALL.19, 20, 21

In pediatric AML, flow cytometry has been widely used for correlative studies of MRD and outcome.22, 23, 24, 25 In the St Jude AML02 study, MRD remained a predictive prognostic factor despite treatment intensification for MRD-positive patients.4 The prognostic significance of MRD was also demonstrated in adult patients with AML.26, 27 Maurillo et al.28 found that levels of MRD after consolidation therapy were particularly informative: MRD-negative patients had a significantly more favorable outcome, regardless of whether they received autologous or allogeneic stem cell transplantation. In summary, evidence for the prognostic importance of MRD in both ALL and AML is extraordinarily strong; arguably, it is more compelling than that supporting any other prognostic factors used in the past for risk stratification.

The prevalence of MRD varies among different genetic subtypes of ALL and AML;3, 4, 29, 30, 31 it is associated with gene profiles of leukemic cells,32, 33 as well as germline- or leukemia-associated gene polymorphisms.34 Nevertheless, the predictive information provided by MRD testing is independent of these associations, as MRD measurements reflect the composite influence of multiple factors, including variables related to drug interactions, dosage, pharmacokinetics and compliance. Moreover, besides its use for prognostication, MRD monitoring can also recognize leukemia relapse before it is morphologically overt, assess the leukemia burden before transplant and measure the efficacy of a treatment regimen in relation to that of its predecessor. Therefore, it is likely that MRD measurements will be increasingly used in risk stratification algorithms for patients with acute leukemia, and as an end point for the assessment of novel therapeutic agents. The widening application of MRD testing is supported by novel methodologies that promise to further improve its sensitivity and reliability.35, 36, 37

Acknowledgments

This work was supported by grants from the National Medical Research Council of Singapore, the National Cancer Institute and by the American Lebanese Syrian Associated Charities (ALSAC).

This article was published as part of a supplement that was supported by Novartis, MSD Italia, Roche, Celgene, GlaxoSmithKline, Sanofi, Gilead, Adienne, Italfarmaco, Pierre Fabre Pharmaceuticals with an unrestricted educational contribution to AREO—Associazione Ricerche Emato-Oncologiche (Genoa) and AMS—Associazione Malattie del Sangue (Milan) for the purpose of advancing research in acute and chronic leukemia.

The author declares no conflict of interest.

References

  1. Bruggemann M, Schrauder A, Raff T, Pfeifer H, Dworzak M, Ottmann OG et al. Standardized MRD quantification in European ALL trials: proceedings of the Second International Symposium on MRD assessment in Kiel, Germany, 18–20 September 2008. Leukemia 2010; 24: 521–535. [DOI] [PubMed] [Google Scholar]
  2. Shook D, Coustan-Smith E, Ribeiro RC, Rubnitz JE, Campana D. Minimal residual disease quantitation in acute myeloid leukemia. Clin Lymphoma Myeloma 2009; 9 (Suppl 3): S281–S285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Pui CH, Campana D, Pei D, Bowman WP, Sandlund JT, Kaste SC et al. Treating childhood acute lymphoblastic leukemia without cranial irradiation. N Engl J Med 2009; 360: 2730–2741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Rubnitz JE, Inaba H, Dahl GV, Ribeiro RC, Bowman WP, Taub JW et al. Minimal residual disease-directed therapy for childhood acute myeloid leukemia: results of the AML02 multicenter trial. Lancet Oncol 2010; 11: 543–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Coustan-Smith E, Sancho J, Hancock ML, Boyett JM, Behm FG, Raimondi SC et al. Clinical importance of minimal residual disease in childhood acute lymphoblastic leukemia. Blood 2000; 96: 2691–2696. [PubMed] [Google Scholar]
  6. Borowitz MJ, Devidas M, Hunger SP, Bowman WP, Carroll AJ, Carroll WL et al. 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: 5477–5485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Conter V, Bartram CR, Valsecchi MG, Schrauder A, Panzer-Grumayer R, Moricke A et al. Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood 2010; 115: 3206–3214. [DOI] [PubMed] [Google Scholar]
  8. Pui CH, Pei D, Campana D, Bowman WP, Sandlund JT, Kaste SC et al. Improved prognosis for older adolescents with acute lymphoblastic leukemia. J Clin Oncol 2011; 29: 386–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. van der Velden V, Corral L, Valsecchi MG, Jansen MW, De LP, Cazzaniga G et al. Prognostic significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol. Leukemia 2009; 23: 1073–1079. [DOI] [PubMed] [Google Scholar]
  10. Schrappe M, Valsecchi MG, Bartram CR, Schrauder A, Panzer-Grumayer R, Moricke A et al. Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: results of the AIEOP-BFM-ALL 2000 study. Blood 2011; 118: 2077–2084. [DOI] [PubMed] [Google Scholar]
  11. Coustan-Smith E, Sancho J, Behm FG, Hancock ML, Razzouk BI, Ribeiro RC et al. Prognostic importance of measuring early clearance of leukemic cells by flow cytometry in childhood acute lymphoblastic leukemia. Blood 2002; 100: 52–58. [DOI] [PubMed] [Google Scholar]
  12. Basso G, Veltroni M, Valsecchi MG, Dworzak MN, Ratei R, Silvestri D 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: 5168–5174. [DOI] [PubMed] [Google Scholar]
  13. Stow P, Key L, Chen X, Pan Q, Neale GA, Coustan-Smith E et al. Clinical significance of low levels of minimal residual disease at the end of remission induction therapy in childhood acute lymphoblastic leukemia. Blood 2010; 115: 4657–4663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Raetz EA, Borowitz MJ, Devidas M, Linda SB, Hunger SP, Winick NJ et al. Reinduction platform for children with first marrow relapse of acute lymphoblastic Leukemia: A Children's Oncology Group Study. J Clin Oncol 2008; 26: 3971–3978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bader P, Kreyenberg H, Henze GH, Eckert C, Reising M, Willasch A et al. Prognostic value of minimal residual disease quantification before allogeneic stem-cell transplantation in relapsed childhood acute lymphoblastic leukemia: the ALL-REZ BFM Study Group. J Clin Oncol 2009; 27: 377–384. [DOI] [PubMed] [Google Scholar]
  16. Leung W, Campana D, Yang J, Pei D, Coustan-Smith E, Gan K et al. High success rate of hematopoietic cell transplantation regardless of donor source in children with very high-risk leukemia. Blood 2011; 118: 223–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bruggemann M, Raff T, Flohr T, Gokbuget N, Nakao M, Droese J et al. Clinical significance of minimal residual disease quantification in adult patients with standard-risk acute lymphoblastic leukemia. Blood 2006; 107: 1116–1123. [DOI] [PubMed] [Google Scholar]
  18. Raff T, Gokbuget N, Luschen S, Reutzel R, Ritgen M, Irmer S et al. Molecular relapse in adult standard-risk ALL patients detected by prospective MRD monitoring during and after maintenance treatment: data from the GMALL 06/99 and 07/03 trials. Blood 2007; 109: 910–915. [DOI] [PubMed] [Google Scholar]
  19. Bassan R, Spinelli O, Oldani E, Intermesoli T, Tosi M, Peruta B et al. Improved risk classification for risk-specific therapy based on the molecular study of MRD in adult ALL. Blood 2009; 113: 4153–4162. [DOI] [PubMed] [Google Scholar]
  20. Patel B, Rai L, Buck G, Richards SM, Mortuza Y, Mitchell W et al. Minimal residual disease is a significant predictor of treatment failure in non T-lineage adult acute lymphoblastic leukaemia: final results of the international trial UKALL XII/ECOG2993. Br J Haematol 2010; 148: 80–89. [DOI] [PubMed] [Google Scholar]
  21. Giebel S, Stella-Holowiecka B, Krawczyk-Kulis M, Gokbuget N, Hoelzer D, Doubek M et al. Status of minimal residual disease determines outcome of autologous hematopoietic SCT in adult ALL. Bone Marrow Transplant 2009; 45: 1095–1101. [DOI] [PubMed] [Google Scholar]
  22. Sievers EL, Lange BJ, Alonzo TA, Gerbing RB, Bernstein ID, Smith FO et al. Immunophenotypic evidence of leukemia after induction therapy predicts relapse: results from a prospective Children's Cancer Group study of 252 acute myeloid leukemia patients. Blood 2003; 101: 3398–3406. [DOI] [PubMed] [Google Scholar]
  23. Coustan-Smith E, Ribeiro RC, Rubnitz JE, Razzouk BI, Pui CH, Pounds S et al. Clinical significance of residual disease during treatment in childhood acute myeloid leukemia. Br J Haematol 2003; 123: 243–252. [DOI] [PubMed] [Google Scholar]
  24. Langebrake C, Creutzig U, Dworzak M, Hrusak O, Mejstrikova E, Griesinger F et al. Residual disease monitoring in childhood acute myeloid leukemia by multiparameter flow cytometry: the MRD-AML-BFM Study Group. J Clin Oncol 2006; 24: 3686–3692. [DOI] [PubMed] [Google Scholar]
  25. van der Velden VH, Sluijs-Geling A, Gibson BE, te Marvelde JG, Hoogeveen PG, Hop WC et al. Clinical significance of flowcytometric minimal residual disease detection in pediatric acute myeloid leukemia patients treated according to the DCOG ANLL97/MRC AML12 protocol. Leukemia 2010; 24: 1599–1606. [DOI] [PubMed] [Google Scholar]
  26. San Miguel JF, Vidriales MB, Lopez-Berges C, Diaz-Mediavilla J, Gutierrez N, Canizo C et al. Early immunophenotypical evaluation of minimal residual disease in acute myeloid leukemia identifies different patient risk groups and may contribute to postinduction treatment stratification. Blood 2001; 98: 1746–1751. [DOI] [PubMed] [Google Scholar]
  27. Kern W, Voskova D, Schoch C, Hiddemann W, Schnittger S, Haferlach T. Determination of relapse risk based on assessment of minimal residual disease during complete remission by multiparameter flow cytometry in unselected patients with acute myeloid leukemia. Blood 2004; 104: 3078–3085. [DOI] [PubMed] [Google Scholar]
  28. Maurillo L, Buccisano F, Del Principe MI, Del PG, Spagnoli A, Panetta P et al. Toward optimization of postremission therapy for residual disease-positive patients with acute myeloid leukemia. J Clin Oncol 2008; 26: 4944–4951. [DOI] [PubMed] [Google Scholar]
  29. Campana D. Minimal residual disease in acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program 2010; 2010: 7–12. [DOI] [PubMed] [Google Scholar]
  30. Coustan-Smith E, Mullighan CG, Onciu M, Behm FG, Raimondi SC, Pei D et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol 2009; 10: 147–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Mullighan CG, Su X, Zhang J, Radtke I, Phillips LA, Miller CB et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med 2009; 360: 470–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Cario G, Stanulla M, Fine BM, Teuffel O, Neuhoff NV, Schrauder A et al. Distinct gene expression profiles determine molecular treatment response in childhood acute lymphoblastic leukemia. Blood 2005; 105: 821–826. [DOI] [PubMed] [Google Scholar]
  33. Flotho C, Coustan-Smith E, Pei D, Cheng C, Song G, Pui CH et al. A set of genes that regulate cell proliferation predicts treatment outcome in childhood acute lymphoblastic leukemia. Blood 2007; 110: 1271–1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Yang JJ, Cheng C, Yang W, Pei D, Cao X, Fan Y et al. Genome-wide interrogation of germline genetic variation associated with treatment response in childhood acute lymphoblastic leukemia. JAMA 2009; 301: 393–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Coustan-Smith E, Song G, Clark C, Key L, Liu P, Mehrpooya M et al. New markers for minimal residual disease detection in acute lymphoblastic leukemia. Blood 2011; 117: 6267–6276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Pedreira CE, Costa ES, Almeida J, Fernandez C, Quijano S, Flores J et al. A probabilistic approach for the evaluation of minimal residual disease by multiparameter flow cytometry in leukemic B-cell chronic lymphoproliferative disorders. Cytometry A 2008; 73A: 1141–1150. [DOI] [PubMed] [Google Scholar]
  37. Faham M, Willis T, Moorhead M, Carlton V, Zheng J, Campana D. Highly sensitive detection of minimal residual disease in acute lymphoblastic leukemia using next-generation sequencing of immunoglobulin heavy chain variable region. Blood (ASH Annual Meeting Abstracts) 2011; 118: 2540. [Google Scholar]

Articles from Leukemia Supplements are provided here courtesy of Nature Publishing Group

RESOURCES