SciELO - Scientific Electronic Library Online

 
vol.77 número6SIRS o no SIRS: ¿es esa la infección? Una revisión crítica de los criterios de definición de sepsis índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • No hay artículos similaresSimilares en SciELO

Compartir


Boletín médico del Hospital Infantil de México

versión impresa ISSN 1665-1146

Bol. Med. Hosp. Infant. Mex. vol.77 no.6 México nov./dic. 2020  Epub 27-Nov-2020

https://doi.org/10.24875/bmhim.20000171 

Artículo de revisión

Aberrant immunophenotypes in acute lymphoblastic leukemia

Inmunofenotipos aberrantes en la leucemia linfoblástica aguda

Miguel E. Cuéllar-Mendoza1 

Francisco R. Chávez-Sánchez1 

Elisa Dorantes-Acosta2 

Blanca M. Arsuaga-Jiménez3 

Marta Zapata-Tarrés3  * 

1Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico

2Departamento de Hemato-Oncología, Hospital Infantil de México Federico Gómez, Mexico

3Departamento de Oncología, Instituto Nacional de Pediatría. Mexico City, Mexico


Abstract

Acute lymphoblastic leukemia (ALL) is the most prevalent hematologic neoplasia worldwide. To classify leukemia, we analyzed the immunophenotypic characteristics in the neoplastic cells obtained with antibodies by cell flow cytometry or immunohistochemistry. The aberrant immunophenotypes are antigen expression patterns that differ from the normal hematopoietic maturation process, which can include some different lineage antigens such as myeloid antigens in ALL or asynchronous expression of antigens. These aberrant immunophenotypes have been studied as prognostic factors and residual disease markers. In this review, some aspects of aberrant immunophenotypes are addressed, including definition, epidemiology, and potential uses.

Key words Immunophenotypes; Leukemia; Lymphoblastic; Hematologic; Review

Resumen

La leucemia linfoblástica aguda es la neoplasia hematológica más prevalente en el mundo. Para clasificar la leucemia se utilizan características inmunofenotípicas en las células neoplásicas que se pueden observar con anticuerpos en la citometría de flujo o mediante inmunohistoquímica. Los inmunofenotipos aberrantes son los patrones de expresión de los antígenos que difieren del proceso normal de maduración hematopoyética, y pueden incluir antígenos de linajes diferentes, como antígenos mieloides en la leucemia linfoblástica aguda, o la expresión asincrónica de antígenos. Estos inmunofenotipos aberrantes se han estudiado como factores de pronóstico y como marcadores de enfermedad mínima residual. En esta revisión se abordan algunos aspectos de los inmunofenotipos aberrantes, incluyendo su definición, epidemiología y usos potenciales.

Palabras clave Inmunofenotipos; Leucemia; Linfoblástica; Hematología; Revisión

Introduction

Acute lymphoblastic leukemia (ALL) is a malignant transformation of hematopoietic progenitor cells in the blood marrow, blood, and extramedullary sites, characterized by the accumulation of altered hematopoietic progenitors in those sites1. ALL is the most common malignancy in childhood2,3. In the United States, it has a global incidence of 1.7 cases/100,000 inhabitants4, while in middle-income countries like Mexico, the incidence is higher: 7.98/100,000 inhabitants5. The first classification of ALL was based on the morphological criteria of the French-American-British classification, which includes the nucleus/cytoplasm ratio, nucleoli, size of the cell, vacuoles6, and the fact that the bone marrow must contain at least 20% of blast cells (Table 1)7. After the introduction of flow cytometry in clinical diagnosis, lineage markers to classify leukemia as B cell origin or T cell origin have been widely used. This method revealed that some abnormal antigens can appear in the neoplastic cells.

Table 1 French-American-British classification for acute lymphoblastic leukemia46 

Type Morphology Immunophenotype
L1 Homogeneous blasts, regular nucleus, homogenous chromatin, small or inexistent nucleoli, mild basophilia B cell: CD19, CD22, CD79a, CD10, CD20, Ig cytoplasmic, or superficial T cell: CD3, CD7, CD5, CD2, or CD4
L2 Irregular nucleus, heterogeneous chromatin, and large nucleoli B cell: CD19, CD22, CD79a, CD10, CD20, Ig cytoplasmic, or superficial T cell: CD3, CD7, CD5, CD2, or CD4
L3 Large blasts, prominent nucleoli, abundant cytoplasm, vacuolation covering the nucleus B cell: CD19, CD22, CD79a, CD10, CD20, Ig cytoplasmic, or superficial T cell: CD3, CD7, CD5, CD2, or CD4

Ig: immunoglobulin.

Common immunophenotype markers in ALL

At present, leukemia can be classified into B cell or T cell types, depending on the markers that the neoplastic cells express in a flow cytometry assay. The most common cellular markers are immunoglobulins (Ig): CD19 or CD22 for B cell neoplasia, and CD3 and T cell receptor (TCR) for T cell neoplasia (Table 2)8,9. These markers are present in the non-neoplastic counterpart and represent well-characterized molecules that are typical for these lineages. Even in the European Group for Immunological Characterization of Acute Leukemia (EGIL) first proposal for the classification of acute leukemia, it was recognized that some of the ALL express myeloid antigens9, which are not present in their normal counterpart cells. Further studies defined biphenotypic and bilineal leukemia. At present, in Mexico, some efforts have been made to establish an immunological classification for acute leukemia (Table 3).

Table 2 Markers used for lineage classification of acute lymphoblastic leukemia 

Lineage Markers
B cell Surface immunoglobulins, cytoplasmic light chains, CD19, CD22, CD79
T cell CD3 (cytoplasmic or membrane), CD2, CD5, CD8, T cell receptor

Table 3 Markers proposed in the National Consensus of Acute Leukemia Phenotyping in Mexico47 

Type of ALL Lineage Differentiation/Maturation Subclassification Optional
B cell precursor CD19 Cytoplasmic CD79 CD34, CD45, CD20, CD38 CD10, Ig superficial, µ cytoplasmic chains TdT, CD13, CD33
T cell precursor CD3, CD7 CD34, CD45, TdT, CD1a, CD99 — CD2, CD4, CD5, CD8, CD13, CD33

ALL: acute lymphoblastic leukemia; TdT: terminal deoxynucleotidyl transferase; Ig: immunoglobulin.

Khalidi et al. identified non-significant differences between primary and relapsed ALL patients10.

Biphenotypic and bilineal leukemia

EGIL defines biphenotypic leukemia as a group in which blasts express simultaneously myeloid and lymphoid antigens, with a score of ≥ 2 points present in two different lineages (Table 4)11.

Table 4 EGIL criteria for biphenotypic leukemia 

Points B cell lineage T cell lineage Myeloid lineage
2 CD7, CD22, IgM CD3, TCR MPO
1 CD19, CD20, CD10 CD2, CD5, CD8, CD10 CD117, CD13, CD33
0.5 TdT y CD24 TdT, CD7, CD1a CD14, CD15, CD64

*For the diagnostic of biphenotypic leukemia, myeloperoxidase (a major myeloid antigen), and another major antigen of B or T cell origin should be present. The diagnostic of aberrant immunophenotypes is determined with one point in two different categories.EGIL: European Group for Immunological Characterization of Acute Leukemia; MPO: myeloperoxidase; TCR: T cell receptor; TdT: terminal deoxynucleotidyl transferase.

Bilineal leukemia is defined as a heterogeneous group of hematopoietic malignancies, with blasts that cannot be classified as myeloid or lymphoid, or blasts from both lineages12.

Not only EGIL, but the World Health Organization also proposed criteria for mixed-phenotype blasts in the Classification Tumors of Hematopoietic and Lymphoid Tissues 2016 update (Table 5)11-13. This classification comprises bilineal and biphenotypic leukemia into this entity and includes 2-5% of all leukemia diagnosis. No threshold level of antigen expression is defined for the diagnosis14.

Table 5 WHO criteria for mixed-phenotype blasts 

Lineage Markers
Myeloid Myeloperoxidase or two of the following: CD11c, non-specific esterase, CD14, CD64, lysozyme
T lineage Cytoplasmic or surface CD3
B lineage Strong CD19 plus at least one of the following: CD79a, cytoplasmic CD22 or CD10; or weak CD19 and two of the following: CD79a, cytoplasmic CD22, or CD10

WHO: World Health Organization.

Aberrant immunophenotypes in ALL

Some leukemias show antigen expression of two cellular lineages but not all the biphenotypic, bilineal, or mixed phenotype criteria. The expression of these antigens has been termed anomalous or aberrant15. In B cell ALL, myeloid antigens are the best-studied antigens. Another definition of aberrant immunophenotypes is the antigen expression patterns in neoplastic cells that differ from the normal antigens in the maturation process of hematopoietic cells. In essence, this includes other lineage antigens: myeloid antigens in ALL, B cell antigens in T cell ALL or vice versa, or antigen asynchrony (when earlier phase antigens are expressed in mature cells). The threshold for positivity10,16 is 20-30% of the isolated leukemic cells17.

Conflicting information on the prevalence of aberrant antigens has been reported. These differences can be explained by the variation of antibodies against antigens, diverse cutoff levels, or the samples used in the analysis18. Seegmiller et al. reported 86.5% of myeloid antigens in 200 cases of B lineage ALL and 9% of T cell antigens in their series19. The Brazilian study by Lopes et al. showed only 49.2% of myeloid expression in T and B cell ALL20. In turn, Bachir et al. studied a population of Moroccan children and found myeloid expression in 46.3% of B cell ALL and 22% of T cell ALL21; other studies mentioned higher prevalence in T cell ALL22. Ibrahim et al. mentioned a percentage as low as 5% of myeloid expression18. From these reports, it is assumed that incidence varies depending on the population studied. The gathered information is consistent in the fact that the expression of myeloid antigens is higher in B cell ALL. Furthermore, Khalidi et al. reported T cell markers in B cell ALL, CD3, CD4, and CD7, were expressed in < 5% of the cases10. B cell markers in T cell ALL were expressed in 15% of the cases in the Sharma study, with CD79a and CD19 being the most commonly found23. Bhushan et al. reported a higher incidence of myeloid antigens in adults24. However, these findings were not observed in other studies10,19. In Argentinian patients, Novoa et al. characterized the presence of antigen asynchrony in B cell ALL, with CD34+ CD20+ and CD34+ CD10- being the most frequently expressed phenotypes25. The differences found in these studies reported during the past 20 years were the main reason to do a review on this topic. Table 6 shows the studies and results from different authors15,16,18,26-28.

Table 6 Several studies of myeloid antigens as possible prognostic factors in patients with acute lymphoblastic leukemia 

Author Year Type of leukemia (B, T or both) Results
Tabernero et al.26 2001 B 13 cases with BCR/ABL (Abelson) translocation were positive for CD13
Yeneral et al.16 2002 Both The patients positive to CD13 had better overall survival and disease-free survival (85 vs. 50% and 67 vs. 43%, respectively)
Vitale et al.27 2006 Both No correlation was found between the expression of CD13 and the disease-free survival or overall survival
Noronha et al.15 2011 Both The positive myeloid group achieved more remission cases
Supriyadi et al.28 2012 Both The negative myeloid group got a better disease-free survival compared with the positive myeloid group (80 vs. 67%)
Ibrahim et al.18 2017 Both CD13 positive group had complete remission and fewer blasts in bone marrow, compared to the negative group

BCR: B cell receptor.

One of the most frequent aberrant myeloid markers is CD13, a glycoprotein with peptidase activity18 expressed in normal hematopoietic cells in the processes of maturation, from myeloblasts to granulocytes. In the Seegmiller study, CD13 was the most common myeloid antigen present in ALL (54.5%)19. Another antigen, CD33, is a transmembrane protein present in myeloblast with inhibitory functions, expressed in promyelocytes, monocytes, and erythrocyte29. CD33 was found in 43% in the Seegmiller study. Other myeloid markers such as CD117 are not frequently expressed in ALL30,31. HLA-DR is more frequently found in B cell precursors ALL than in T cell ALL. NK markers, like CD56, have been found on B cell ALL in low percentages10.

Aberrant immunophenotypes as a prognostic factor

At present, several contradictory results from different studies on this topic have emerged. In 2017, Kavianpour et al.30 discussed some studies that found a lower death rate in the positive myeloid group. However, the results of other authors are not consistent with this study16,24,27,31. Copeland and McGuire addressed the same problem32.

The Children’s Cancer Group reported that the aberrant immunophenotype expression was not an adverse prognostic factor: 1557 children enrolled with B or T cell ALL, but only 13.9% and 2.8%, respectively, expressed positive myeloid antigens17. The research of the St Jude Children’s Research Hospital in acute mixed lineage leukemia stated that lineage infidelity does not have prognostic significance33. Moreover, The Medical Research Council, in the United Kingdom, showed no prognostic significance of the expression of CD13 and CD3334. Lopez et al. found a significantly higher platelet value in those patients with positive myeloid markers20. In adult patients, Yenerel et al. found differences in overall survival between the myeloid positive and negative groups (85 vs. 50%); also, the disease-free survival was different (67 vs. 43%), which was not consistent with previous studies16. In Indonesian patients treated with the ALL 2006 protocol, Supriyadi et al. found that leukemia-free survival analyses showed worse prognosis in the positive myeloid group: disease-free survival was 67% in the positive myeloid group versus 80% in negative myeloid patients28. Sharma et al. reported a lower number of peripheral blasts and lower white blood cell count in the myeloid positive group23, while Amirghofran et al. reported less significance in survival time in CD13 positive cases versus negative cases35. Similar results were found in the Mexican study by Rodriguez et al., as well as a lower disease-free survival period36. Craddock et al. showed an increased relapse risk in CD13 positive cases37, while Dalal et al. proposed a more aggressive treatment for ALL with this immunophenotype38.

Myeloid expression was observed in Philadelphia chromosome-positive ALL, both in children and adults. This chromosomic aberration is considered as bad prognostic in adults. Other chromosomic abnormalities with an expression of myeloid antigens reported by Khalidi et al. were the deletion of band 11q23 and translocations involving this chromosome10. The expression of CD5 in B cell ALL has been reported in association with small supernumerary marker chromosomes39.

Immunophenotypic detection of minimal residual disease

Over the past decades, a considerable effort has been made in developing methods to detect residual leukemia in patients that had achieved clinical remission. Minimal residual disease (MRD) is defined as the detection of at least 0.01% leukemic blasts per mononuclear or total nucleated cells40. The methods for MRD detection are the polymerase chain reaction with fusion gene transcripts or Ig/TCR gene rearrangements by flow cytometry41,42.

In 1990, Campana et al. studied the asynchronous expression and lineage infidelity and detected minimal disease (35%) and operationally leukemia-specific. Only in two of 35 cases, the characteristics in the cells changed43. In 1997, San Miguel et al. reported that immunophenotypic aberrancies were useful in predicting relapse and MRD in acute myeloid leukemia44.

García-Vela et al. characterized four different types of aberrant immunophenotypes or leukemia-associated immunophenotypes that can be detected by flow cytometry, which include lineage infidelity, asynchronous antigen expression, ectopic antigen expression (expression of markers outside the thymus), and antigen overexpression. Therefore, the study of these antigens was suggested for MRD, as they found a high frequency of aberrancies45.

In 1999, Lucio et al. performed multicentric studies to detect MRD with flow cytometry, with positive results detecting malignant lymphocytes from normal lymphoblasts46. Several subsequent studies have demonstrated the same results47,48. Gaipa et al. mentioned that leukemia-associated immunophenotypes are widely applicable, rapid, and direct but operator-dependent and require standardization42. Campana emphasizes the use of large antibody panels and interpretation expertise41. At present, six to eight-color flow cytometry methods are used for leukemia immunophenotyping in MRD42. This approach was useful in the Dhahran Health Center study, where they identified six MRD positive cases in B ALL40. The Euroflow group is recently standardizing new leukemia immunophenotyping and MRD protocols, with a more significant number of immunological markers.

Aberrant immunophenotypes are antigens that differ from the typical expression pattern in hematolymphoid neoplastic cells. The incidence of this phenomenon varies from 5% to 86.5%. This difference could be explained by several reasons. For example, in the study that showed an incidence of 86.5%, several antigens that are generally not measured were considered. Conversely, in the series that considered the two most common myeloid antigens (CD13 and CD33), the percentage was much lower (30-50%). Furthermore, the different laboratories or dilutions used for the antibodies, as well as the positivity threshold that some of the antigens showed in different studies, ranging from 20 to 30%, should be considered. Another crucial variable is the different populations in the studies.

The potential role of aberrant immunophenotypes as a prognostic factor is still controversial. Some studies in low- and medium-income countries reveal that prognosis is different between myeloid positive and myeloid negative ALL. Contrastingly, in more prominent studies from the USA and UK, no significant differences were reported. In an Indonesian study, a different treatment protocol from the St. Jude protocol was used, which could explain the positive results in the evaluation of the disease remission. In patients with aggressive protocols, no evidence that the aberrant immunophenotypes change the prognostic of patients has been found. Other factors that need to be considered are the controversies between different populations and between low- and high-income countries.

In MRD, the detection of different antigens in the cell and the introduction of flow cytometry have been useful for diagnosis and treatment. However, as the panels are extensive, standardization is essential because they are operator-dependent.

Acknowledgments

To the Instituto Nacional de Pediatría and the Hospital Infantil de México Federico Gómez. The authors would also like to thank the PhD program in Medical, Odontological and Health Sciences from UNAM and the grant from CONACyT.

References

1. Terwilliger T, Abdul-Hay M. Acute lymphoblastic leukemia:a comprehensive review and 2017 update. Blood Cancer J. 2017;7:1-12. [ Links ]

2. Mitchell C, Hall G, Clarke RT. Acute leukaemia in children:diagnosis and management. BMJ. 2009;338:1491-5. [ Links ]

3. Dorantes-Acosta E, Medina-Sanson A, Dávila-Ornelas K, López-Martínez B. Clasificación inmunológica de las leucemias agudas linfoblásticas del hospital infantil de México Federico Gómez, de acuerdo al EGIL (European Group for the Immunological Classification of Leukemia). Gac Mex Oncol. 2013;12:136-42. [ Links ]

4. National Cancer Institute. National Cancer Institute, Surveillance Research Program. Cancer Statistics Review 1975-2013:age-adjusted SEER Incidence and US Death Rates and 5-year Relative Survival Rates. Bethesda, MD:National Cancer Institute;2013. Available from:https://www.seer.cancer.gov/archive/csr/1975_2013/results_single/sect_01_table.05_2pgs.pdf. [ Links ]

5. Rivera-Luna R, Velasco-Hidalgo L, Zapata-Tarrés M, Cárdenas-Cardos R, Aguilar-Ortiz MR. Current outlook of childhood cancer epidemiology in a middle-income country under a public health insurance program. Pediatr Hematol Oncol. 2017;34:43-50. [ Links ]

6. Zafar MN. FAB classification of acute lymphoblastic leukaemia (ALL) and its relevence to all in Karachi children. J Pak Med Assoc. 1985;35:233-6. [ Links ]

7. Chiaretti S, Zini G, Bassan R. Diagnosis and subclassification of acute lymphoblastic leukemia. Med J Hematol Infect Dis. 2014;6:e2014073. [ Links ]

8. Gajl-Peczalska KJ, Bloomfield CD, Nesbit ME, Kersey JH. B-cell markers on lymphoblasts in acute lymphoblastic leukaemia. Clin Exp Immunol. 1974;17:561-9. [ Links ]

9. Bene MC, Castoldi G, Knapp W, Ludwig WD, Matutes E, Orfao A, et al. Proposals for the immunological classification of acute leukemias. Leukemia. 1995;9:1783-6. [ Links ]

10. Khalidi HS, Chang KL, Medeiros LJ, Brynes RK, Slovak ML, Murata-Collins JL, et al. Acute lymphoblastic leukemia:survey of immunophenotype, French-American-British classification, frequency of myeloid antigen expression, and karyotypic abnormalities in 210 pediatric and adult cases. Am J Clin Pathol. 1999;111:467-76. [ Links ]

11. Charles NJ, Boyer DF. Mixed-phenotype acute leukemia:diagnostic criteria and pitfalls. Arch Pathol Lab Med. 2017;141:1462-8. [ Links ]

12. Cernan M, Szotkowski T, Pikalova Z. Mixed-phenotype acute leukemia:state-of-the-art of the diagnosis, classification and treatment. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2017;161:234-41. [ Links ]

13. Arber DA, Orazi A, Hasserjian R, Borowitz MJ, Beau Le MM, Bloomfield CD, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127:2391-406. [ Links ]

14. Khan M, Siddiqi R, Naqvi K. An update on classification, genetics, and clinical approach to mixed phenotype acute leukemia (MPAL). Ann Hematol. 2018;97:945-53. [ Links ]

15. Noronha EP, Marinho HT, Thomaz EB, Silva CA, Veras GL, Oliveira RA. Immunophenotypic characterization of acute leukemia at a public oncology reference center in Maranhao, Northeastern Brazil. Sao Paulo Med J. 2011;129:392-401. [ Links ]

16. Yeneral MN, Atamer T, Yavuz AS, Kucukkaya R, Besisik S, Aktan M, et al. Myeloid antigen expression provides favorable outcome in patients with adult acute lymphoblastic leukemia:a single-center study. Ann Hematol. 2002;81:498-503. [ Links ]

17. Uckun FM, Sather HN, Gaynon PS, Arthur DC, Trigg ME, Tubergen DG, et al. Clinical features and treatment outcome of children with myeloid antigen-positive acute lymphoblastic leukemia:a report from the children's cancer group. Blood. 1997;90:28-35. [ Links ]

18. Ahmed H, Tahir N, Obed F. Cytokines profiling as prognostic markers in newly diagnosed acute myeloid leukemia. Iraqi J Hematol. 2017;6:12-6. [ Links ]

19. Seegmiller AC, Kroft SH, Karandikar NJ, McKenna RW. Characterization of immunophenotypic aberrancies in 200 cases of B acute lymphoblastic leukemia. Am J Clin Pathol. 2009;132:940-9. [ Links ]

20. Lopes TC, Andrade KN, Camelo NL, Rodrigues VP, Oliveira RA. Influence of aberrant myeloid expression on acute lymphoblastic leukemia in children and adolescents from Maranhao, Brazil. Genet Mol Res. 2014;13:10301-7. [ Links ]

21. Bachir F, Bennani S, Lahjouji A, Cherkaoui S, Harif M, Khattab M, et al. Characterization of acute lymphoblastic leukemia subtypes in Moroccan children. Int J Pediatr. 2009;2009:674801. [ Links ]

22. Mazher N, Malik N, Imran A, Chughtai O, Chughtai AS. Aberrant expression of CD markers in acute leukemia. Ann Pak Inst Med Sci. 2013;9:99-102. [ Links ]

23. Sharma M, Sachdeva MS, Varma N, Varma S, Marwaha R. Characterization of immunophenotypic aberrancies in adult and childhood acute lymphoblastic leukemia:a study from Northern India. J Cancer Res Ther. 2016;12:620-6. [ Links ]

24. Bhushan B, Chauhan PS, Saluja S, Verma S, Mishra AK, Siddiqui S, et al. Aberrant phenotypes in childhood and adult acute leukemia and its association with adverse prognostic factors and clinical outcome. Clin Exp Med. 2010;10:33-40. [ Links ]

25. Novoa V, Nuñez NA, Carballo OG, Lessa CF. Inmunofenotipos aberrantes en leucemias agudas en una población hospitalaria de Buenos Aires. Medicina. 2013;73:9-16. [ Links ]

26. Tabernero MD, Bortoluci AM, Alaejos I, López-Berges MC, Rasillo A, García-Sanz R, et al. Adult precursor B-ALL with BCR/ABL gene rearrangements displays a unique immunophenotype based on the pattern of CD10, CD34, CD13 and CD38 expression. Leukemia. 2001;15:406-14. [ Links ]

27. Vitale A, Guarini A, Ariola C, Meloni G, Perbellini O, Pizzuti M, et al. Absence of prognostic impact of CD13 and/or CD33 antigen expression in adult acute lymphoblastic leukemia. Results of the GIMEMA ALL 0496 trial. Haematologica. 2007;92:342-8. [ Links ]

28. Supriyadi E, Veerman AJ, Sutaryo S, Purwanto I, Vd Ven PM, Cloos J. Myeloid antigen expression in childhood acute lymphoblastic leukemia and its relevance for clinical outcome in Indonesian ALL-2006 protocol. J Oncol. 2012;2012:1-7. [ Links ]

29. Sato TN, Hayashi M. On the origin of species specificity of fibronectin. Immunological identification of a species-specific domain of human fibronectin. J Biochem. 1986;99:653-61. [ Links ]

30. Kavianpour M, Ketabchi N, Saki N. Prognostic significance of aberrant expression of CD markers in acute lymphoblastic leukemia. Mag Eur Med Oncol. 2017;10:164-9. [ Links ]

31. Suggs JL, Cruse JM, Lewis RE. Aberrant myeloid marker expression in precursor B-cell and T-cell leukemias. Exp Mol Pathol. 2007;83:471-3. [ Links ]

32. Copeland E, McGuire E. The biology and treatment of acute lymphoblastic leukemia in adults. Blood. 1995;85:1151-68. [ Links ]

33. Rubnitz JE, Onciu M, Pounds S, Shurtleff S, Cao X, Raimondi SC, et al. Acute mixed lineage leukemia in children:the experience of St Jude children's research hospital. Blood. 2009;113:5083-9. [ Links ]

34. Hann IM, Richards SM, Eden OB, Hill FG. Analysis of the immunophenotype of children treated on the medical research council United Kingdom acute lymphoblastic leukaemia trial XI (MRC UKALLXI). Medical research council childhood leukaemia working party. Leukemia. 1998;12:1249-55. [ Links ]

35. Amirghofran Z, Shamseddin A, Shahriari M. Prognostic value of myeloid antigen expression in acute lymphoblastic leukemia. Med J Islam Repub Iran. 2000;14:111-4. [ Links ]

36. Rodríguez-Rodríguez S, Pomerantz A, Demichelis-Gómez R, Barrera-Lumbreras G, Barrales-Benítez OV, Lopez-Karpovitch X, et al. Impact of aberrant antigens in the outcome of patients with acute leukemia at a referral institution in Mexico City. Rev Invest Clin. 2016;68:305-13. [ Links ]

37. Craddock KJ, Chen Y, Brandwein JM, Chang H. CD13 expression is an independent adverse prognostic factor in adults with Philadelphia chromosome-negative B cell acute lymphoblastic leukemia. Leuk Res. 2013;37:759-64. [ Links ]

38. Dalal BI, Al Mugairi A, Pi S, Lee SY, Khare NS, Pal J, et al. Aberrant expression of CD13 identifies a subgroup of standard-risk adult acute lymphoblastic leukemia with inferior survival. Clin Lymphoma Myeloma Leuk. 2014;14:239-44. [ Links ]

39. Mutreja D, Pati HP, Bansal D, Sharma RK, Jain S. Aberrant immunophenotypic expression of CD5 in a case of b acute lymphoblastic leukemia:a case report. Indian J Hematol Blood Transfus. 2014;30:212-4. [ Links ]

40. Amra N, Sheikh S, Abushullaih B, Al-Faris N, Al-Khatti A, Al-Sayed H. Minimal residual disease program for acute lymphoblastic leukemia at Dhahran health center. J Appl Hematol. 2016;7:17-23. [ Links ]

41. Campana D. Minimal residual disease in acute lymphoblastic leukemia. Semin Haematol. 2009;46:100-6. [ Links ]

42. Gaipa G, Basso G, Biondi A, Campana D. Detection of minimal residual disease in pediatric acute lymphoblastic leukemia. Cytometry B Clin Cytom. 2013;84:359-69. [ Links ]

43. Campana D, Coustan-Smith E, Janossy G. The immunologic detection of minimal residual disease in acute leukemia. Blood. 1990;76:163-71. [ Links ]

44. San Miguel JF, Martínez A, Macedo A, Vidriales MB, López-Berges C, González M, et al. Immunophenotyping investigation of minimal residual disease is a useful approach for predicting relapse in acute myeloid leukemia patients. Blood. 1997;90:2465-70. [ Links ]

45. Vela JA, Monteserin MC, Delgado I, Benito L, Oña F. Aberrant immunophenotypes detected by flow cytometry in acute lymphoblastic leukemia. Leuk Lymphoma. 2000;36:275-84. [ Links ]

46. Lúcio P, Parreira A, Van Den Beemd MW, Van Lochem EG, Van Wering ER, Baars E, 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;13:419-27. [ Links ]

47. Ladines-Castro W, Barragán-Ibañez G, Luna-Pérez MA, Santoyo-Sánchez A, Collazo-Jaloma J, Mendoza-García E, et al. Morphology of leukaemias. Rev Med Hosp Gen Mex. 2015;79:107-13. [ Links ]

48. Arriaga-Pizano L, Ramírez-Ramírez D, Prieto-Chávez J, Pelayo R, Ruiz-Argüelles A. Reporte de la primera reunión nacional de consenso para la inmunofenotipificación de leucemias agudas. Gac Med Mex. 2019;155:20-9. [ Links ]

Ethical disclosures

Protection of human and animal subjects. The authors declare that no experiments were performed on humans or animals for this study.

Confidentiality of data. The authors declare that they have followed the protocols of their work center on the publication of patient data.

Right to privacy and informed consent. The authors declare that no patient identifying information appears in this article.

FundingNone.

Received: October 18, 2019; Accepted: March 11, 2020

* Correspondence: Marta Zapata-Tarrés E-mail: mzapatatarres@gmail.com

Conflicts of interest

The authors declare no conflicts of interest.

Creative Commons License Instituto Nacional de Cardiología Ignacio Chávez. Published by Permanyer. This is an open ccess article under the CC BY-NC-ND license