You are on page 1of 15

Hematology

ISSN: (Print) 1607-8454 (Online) Journal homepage: https://www.tandfonline.com/loi/yhem20

Proteomic tools and new insights for the study of


B-cell precursor acute lymphoblastic leukemia

Alí F. Citalan-Madrid, Griselda A. Cabral-Pacheco, Laura E. Martinez-de-


Villarreal, Laura Villarreal-Martinez, Marisol Ibarra-Ramirez, Idalia Garza-
Veloz, Edith Cardenas-Vargas, Ivan Marino-Martinez & Margarita L. Martinez-
Fierro

To cite this article: Alí F. Citalan-Madrid, Griselda A. Cabral-Pacheco, Laura E. Martinez-de-


Villarreal, Laura Villarreal-Martinez, Marisol Ibarra-Ramirez, Idalia Garza-Veloz, Edith Cardenas-
Vargas, Ivan Marino-Martinez & Margarita L. Martinez-Fierro (2019) Proteomic tools and new
insights for the study of B-cell precursor acute lymphoblastic leukemia, Hematology, 24:1, 637-650,
DOI: 10.1080/16078454.2019.1664127

To link to this article: https://doi.org/10.1080/16078454.2019.1664127

© 2019 The Author(s). Published by Informa Published online: 12 Sep 2019.


UK Limited, trading as Taylor & Francis
Group

Submit your article to this journal Article views: 399

View related articles View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=yhem20
HEMATOLOGY
2019, VOL. 24, NO. 1, 637–650
https://doi.org/10.1080/16078454.2019.1664127

Proteomic tools and new insights for the study of B-cell precursor acute
lymphoblastic leukemia
Alí F. Citalan-Madrida*, Griselda A. Cabral-Pachecoa,b*, Laura E. Martinez-de-Villarrealc, Laura Villarreal-
Martinezd, Marisol Ibarra-Ramirez c, Idalia Garza-Veloz a,b, Edith Cardenas-Vargasa,b,e, Ivan Marino-
Martinezf and Margarita L. Martinez-Fierro a,b
a
Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Zacatecas Autonomous University, Zacatecas,
Mexico; bProgram of Doctorate in Sciences with Orientation in Molecular Medicine, Academic Unit of Human Medicine and Health Sciences,
Zacatecas Autonomous University, Zacatecas, Mexico; cDepartamento de Genetica, Facultad de Medicina, Universidad Autónoma de Nuevo
Leon, Monterrey, Mexico; dHematology Service, Hospital Universitario ‘Dr. José Eleuterio González’, Universidad Autonoma de Nuevo Leon,
Monterrey, Mexico; eHospital General Zacatecas ‘Luz González Cosío’, Zacatecas, Mexico; fDepartamento de Patologia, Facultad de
Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico

ABSTRACT KEYWORDS
B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is a hematological malignancy of Leukemia; haematological
immature B-cell precursors, affecting children more often than adults. The etiology of BCP- malignancy; proteomics;
ALL is still unknown, but environmental factors, sex, race or ethnicity, and genomic acute lymphoblastic
alterations influence the development of the disease. Tools based on protein detection, such leukemia
as flow cytometry, mass spectrometry, mass cytometry and reverse phase protein array,
represent an opportunity to investigate BCP-ALL pathogenesis and to identify new
biomarkers of disease. This review aims to document the recent advancements with respect
to applications of proteomic technologies to study mechanisms of leukemogenesis, how this
information could be used in the discovery of biological targets, and finally we describe the
challenges of application of proteomic tools for the approach of BCP-ALL.

1. Introduction
parameters are commonly used for diagnosis and for
B-cell precursor acute lymphoblastic leukemia (BCP- the establishment of the prognosis of disease.
ALL) is a hematological malignancy characterized by However, because genetic abnormalities and altera-
uncontrolled clonal expansion of B-lymphoid progeni- tions are different in many BCP-ALL cases [9] is necessary
tors in the bone marrow (BM) and extramedullary to count with appropriate tools for the study of the etio-
sites. In childhood, BCP-ALL represents up to 80% of logical basis of the disease and to conduct a risk stratifi-
the total diagnosed cases of acute lymphoblastic leuke- cation for a precise targeted therapy for BCP-ALL.
mias (ALL) [1]. The global incidence of BCP-ALL is 1–5 Among these tools, omics technologies like proteomics
cases per 100,000 people [2], but it differs between have impacted the study of different diseases [10]. In
countries. For example, the incidence of BCP-ALL in BCP-ALL, proteomic tools have awakened the scientific
The United States is 11 cases per 1,000,000 people [3] community attention because the possibility to evalu-
corresponding to 20% for young population with ate the complete proteome in each stage of the
ages between 1 and 20 years (i.e. 3000 new cases per disease and it opens the opportunity to establish their
year) [4], but only 12% of total cases of cancer for functional participation along of the BCP-ALL pro-
adults ≥50 years old, with a peak of incidence at 65 gression and its treatment [11]. At the same time, the
years [5]. In upper middle and middle-income countries implementation of appropriate protocols for molecule
like Mexico, the incidence of ALL is 43.2–44.9 cases per detection, which consider the origin, the collection
1,000,000 people [6] of which, BCP-ALL represents and the correct storage of the sample, is a prerequisite
between 73.2% and 76.1% of total diagnosed cases before to start a proteomics study. In this review, we
with a peak of incidence between 2 and 6 years old will describe the use of proteomic tools in the study of
[6,7]. etiological basis and the discovery of molecular bio-
The etiology of BCP-ALL remains unknown, but the markers. Furthermore, in the context of clinical investi-
important role that genetic factors have in the develop- gation we described the application of proteomic
ment of leukemia has been widely documented [8]. technologies as tools for diagnosis, prognosis, and the
Genetic abnormalities along with immunophenotyping search for new biological targets as well as for monitor-
of leukemic cells as well as blood cells counting ing of minimal residual disease (MRD) of BCP-ALL.

CONTACT Margarita L. Martinez-Fierro margaritamf@uaz.edu.mx; margarita.mtz.fierro@gmail.com


*Equal contribution.
© 2019 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/), which
permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
638 A. F. CITALAN-MADRID ET AL.

2. The proteome and the current tools for its human diseases, to potentiate its utility, and to
evaluation analyze the results obtained by proteomic approaches
is necessary the conjunction with bioinformatic tools.
Proteomics study the structure and function of the pro-
Bioinformatics includes a growing number of data-
teome [12], namely, the complete set of expressed pro-
bases such as NIST/EPA/NIH mass spectral library,
teins. The proteome represents the actual functional
NIST tandem (MS/MS) library (https://chemdata.nist.
state of an organism, whose protein expression is the
gov/), NIST17 (nistmassspeclibrary.com), MS – Sadtler
result of stimuli of different factors both external and
AAFS toxicology section: mass spectra of drugs, and/
internal. Therefore the expressed proteins, are regu-
or SWGDRUG library (http://www.bio-rad.com). These
lated by the metabolism of an individual and at mol-
databases containing information about the structure
ecular level the RNA transcription, alternative splicing,
of proteins, which facilitates the identification of their
and/or post-translational modifications are some of
functions in the development of diseases before any
the processes that modulate that expression. In com-
validation at clinical level (Figure 1). Furthermore, pro-
parison with the relatively steady state of the
teomic tools could represent new opportunities to
genome [13], protein expression at the time that rep-
increase the available information about the etiological
resents the dynamic status of the organism offers an
basis of the disease and to understand how the pro-
unparalleled opportunity to measure and quantify
teome dynamics and changes can influence the
protein changes as a direct indicator of an ongoing
onset of BCP-ALL.
disease process [14]. Proteomic work in patients with
BCP-ALL to date has been carried out mainly with
body fluids such as cerebrospinal fluid (CSF) peripheral
2.1. Flow cytometry as an important tool for
blood (PB) and bone marrow (BM) [15]. The sample
immunophenotyping normal and malignant B-
preparation of body fluids is an important and critical
cells
pre-requisite to achieve efficient data for proteomics
[16]. The sample collection for protein analysis is BCP-ALL originates from B-cell precursors and its detec-
mainly from BM aspirates or blood samples. The lym- tion, classification and the phenotypic features of
phocytes from the samples (BM and PB) can be isolated maturation arrest are mostly based on surface
using Ficoll, while the cells from the CSF can be isolated markers, where FC has become routinely used [19].
by centrifugation. All cells from the three detailed kinds Plastic nature of ALL represents a challenge for the
of samples must be lysated by using kits for human correct early diagnosis of BCP-ALL. Together with cyto-
cells (i.e. RIPA). Protecting proteins from the action of morphology and cytochemistry, FC immunophenotyp-
proteases and other enzymes by degradation is a criti- ing is crucial for the detection of blast cells in suspected
cal consideration for ensuring that analytical results can samples, including the definition of acute leukemias of
be trusted to mirror in vivo state of the proteins or pro- ambiguous lineage. Comparison of the immunopheno-
teomes assayed in tissues-and cell-based samples. The typic features of blasts cells versus normal hematopoie-
time, the reagents used and the storage of the sample tic precursors and immature cells contributes to the
during its processing are key points to avoid the degra- definition of the stage of maturation arrest of the
dation of proteins [15]. The success of this proteomic blast population within the B- and T-lymphoid lineages
research is mainly attributed to the relative ease of as well as the neutrophilic, monocytic, megakaryocytic
sampling and the availability of advanced molecular or erythroid lineages. FC immunophenotyping has also
technology [17]. proven to be of great utility for sensitive detection of
Once the sample has been prepared, the evaluation low levels of residual blast cells and their distinction
of protein expression may be carried out using a series from normal regenerating immature cells in the BM
of high-throughput technologies such as mass spec- of ALL patients during treatment [20–23].
trometry (MS), protein arrays (e.g. RPPA) alone or in Specific phenotypic characterization of ALL blast
combination with other tools like two-dimensional cells is usually performed using lineage associated
polyacrylamide gel electrophoresis (2D-PAGE), and markers such as CD19 for B-cells [24]. Accordingly, in
western blot (WB), whose ultimate goal is to identify B-lineage ALL the most important markers for differen-
and quantify the abundance of proteins under tial diagnosis and subclassification are CD19, CD20,
specified conditions. In the context of BCP-ALL if we CD22, CD24, and CD79a [25]. The combined positive
need to evaluate surface or cytoplasmic protein and negative reaction for these and other complemen-
markers in a living cell, sorting cell tools like flow cyto- tary markers identifies blast cell maturation stage
metry (FC) has been proved to be useful. The obtained which has proven to be of unequivocal clinical value
information can then be used for diagnosis, determi- [26]. Based on classical immunophenotyping, BCP
nation of the cell maturation stage, and the monitoring differentiation has been subdivided into distinct
of MRD during and after BCP-ALL treatment [18]. While stages including pro-B (CD22+CD19−CD10−CD34+),
the technologies mentioned are useful in the study of pre-B-I (CD19+CD10highCD34+cyIgμ−), pre-B-II-Large
HEMATOLOGY 639

Figure 1. Proteomic tools as powerful approach in the investigation of BCP-ALL and the discovery of new molecular biomarkers.
Proteomics involves a series of tools to study the proteome from organisms, tissues, body fluids or cells (left panel). The most
common proteomics tools include mass spectrometry (MS), mass cytometry (CyTOF), two-dimensional polyacrylamide electrophor-
esis (2D-PAGE), flow cytometry, and protein arrays (middle panel). Because of the extensive data produced by proteomics, the use of
bioinformatic tools is necessary for the analysis of hundreds of molecules. Next, analytical and clinical validations are the last steps in
testing the utility of these molecules in the clinical context (right panel).

(CD19+CD10+CD34−cyIgμ+CD20high), pre-B-II-Small present. Surface molecules like CD34 and CD38 com-
(CD19+CD10+CD34−cyIgμ+CD20low) and immature monly aimed at the identification of leukemic-stem
(CD19+CD10+IgK/λ+) stage [27]. cells (LSC) have been observed differentially expressed
The BCP-ALL protocol based in EuroFlow Consor- in some cases of BCP-ALL and rendered as promising
tium panels is designed to distinguish normal or regen- prognostic markers for this malignancy [29].
erating precursor B-cells from BCP-ALL blast cells and For MRD evaluation, the markers suggested are
for full maturation-based classification of BCP-ALL CD10, CD19, CD20, CD34, CD38, CD45, CD73, CD81,
according to EGIL (European Group for the Immuno- CD123 and CD304 [21]. The CD19, CD34, CD45, CD10,
logical Characterization of Leukemias) criteria or WHO CD38, CD20 combination is highly efficient in discrimi-
(World Health Organization) guidelines [21,28]. In the nation of normal/regenerating versus malignant imma-
EuroFlow antibody panel (version 1.10), the markers ture B-cells and could represent an MRD-oriented
included for the BCP-ALL evaluation are CD9, CD10, combination [21,30]. With the advent of new technol-
CD13, CD15, CD19, CD20, CD21, CD22, CD24, CD33, ogies, the panel of new markers has been increasing;
CD34, CD38, CD45, CD58, CD65, CD117, CD123, while some functions of these markers are known,
SmIgK, CyISmIg λ, CyIgμ, SmIgM, NG2 and NuTdT. many of these remain to be studied. Markers currently
From these, the B-lineage-associated markers included used in the clinical practice are CD10, CD19, CD20,
are CD22, CD24, CD10, CD20, CyIg, SmIg. The markers CD22, CD34, CD45, CD123, CyIgμ, SmIgK, CyISmIg λ
with usefulness for differential diagnosis with other and NuTdT [26,31] (Table 1).
acute leukemias include CD13, CD33, CD117, CD15, Notwithstanding the advances in FC BCP-ALL immu-
CD65 (to exclude AML). Other markers suitable for com- nophenotyping and of the accumulated information,
parison with and distinction from normal B-cell devel- there are a growing number of investigations focused
opment patterns are NuTdT, CD10, CD38, CD20 and in to improve the correct distinction and sub-classifi-
CD123 [21]. cation of different stages of B-cell maturation and
The prognosis of the disease is also important and their impact in the prognosis of children and adults
help to apply better treatments to improve the survival with BCP-ALL. On this sense and in order to conduct
rate of leukemic patients. To determine treatment the correct subclassification of BCP-ALL from normal
response and subsequent prognosis in patients lymphoid precursor hematogones, the expression of
treated for BCP-ALL, MRD (by the presence of 0.01% relevant surface markers was analyzed by Sedek et al.
or more ALL cells) measurements should be performed [32]. Multiparameter FC analysis of CD10- and CD10+
[24]. Given that the immunophenotype of leukaemic blast cells showed differential expression of molecules
cells resembles that of BCPs, it is particularly difficult compared to hematogones. However, CD10− blast cells
to detect residual leukaemic cells during therapy inter- were characterized for a higher expression of CD45,
vals, i.e. when regenerating BCPs are abundantly whereas CD10+ blast cells showed increased
640 A. F. CITALAN-MADRID ET AL.

Table 1. Current markers with the clinical application for the classification of BCP-ALL and monitoring of BCP-ALL MRD.
Biomarkers Functions and/or cell type Clinical application in B-ALL References
CD10 c-ALL, lymphatic precursor cells, neutrophils subset of It is the hallmark of childhood most common ALL and of BCP-ALL [21,90]
mature B cells MRD
CD19 B-cell signaling / Precursor B-cells, B-cells Diagnosis of BCP-ALL and subtype classification also it is a marker [21,91]
for BCP-ALL MRD [20,92–95]
CD20 Subpopulation of precursor B-cells, B-cells Marker for mature B-cell differentiation and for BCP-ALL MRD [21,28]
CD22 Regulates B-cell function and migration / Surface It is used for diagnosis of B-ALL and subtype classification. It is [21,96]
expression on B-cells, cytoplasmic expression in precursor strongly associated with the B-lineage, but is not B-cell specific [29,35–38]
B-cells
CD34 Attachment of SC to extracellular matrix in bone marrow / It is used for B-ALL diagnosis and it is a marker of BCP-ALL MRD [21,97]
Myeloid and lymphoid precursor cells [29,98]
CD45 All leukocytes Marker for BCP-ALL MRD [21]
CD123 It is expressed on a subset of B-ALL Marker for BCP-ALL MRD [99]
CyIgμ PreB and B-lymphocytes Marker for pre-B stage of B-III ALL [100]
SmIgκ Surface expression on B-lymphocytes Marker of BCP-ALL [21]
CyISmIgλ Surface expression on B-lymphocytes Marker of BCP-ALL [26]
NuTdT Nuclear expression in lymphoid precursor cells Marker for distinction from normal B-cell development patterns [21]

expression of intranuclear terminal deoxynucleotidyl feasibility of CD73 (in adults) and Neurophilin-1 (NRP-
transferase (TdT), CD22, CD34 and CD20, but a 1)/CD304 (in children) as molecular markers for MRD
decreased expression of CD45 [32]. Even more, FC monitoring in B-ALL, has also been demonstrated
have permitted to discover a significant differential [41]. In this way, FC is a powerful tool for detecting mol-
expression between children and adults diagnosed ecular changes and it could help in the detection of
with BCP-ALL. Seegmiller et al., by using FC analyzed residual malignant cells after therapy. However, it
200 samples from BCP-ALL patients which received a should be kept in mind that, as long as we have
chemotherapy regimen identified 31 different cell- more and better markers, the detection of leukemic
surface markers including B-, myeloid, T-lymphoid cells will be a quicker and easier task, thus strengthen-
and NK-cell antigens [33]. However, a decreased ing the application of FC-based methodologies.
expression of HLA-DR along with a null expression of Additional examples of protein markers with usefulness
CD10 and overexpression of CD45 were more com- in the study and clinical management of BCP-ALL are
monly found in adults than children. These studies described in Tables 2 and 3.
not only demonstrated the heterogeneity and differen-
tial expression in BCP-ALL but also for different age
groups and could partially explain the different out- 2.2. Mass spectrometry in the establishment of
comes observed after the chemotherapy regime. the molecular basis of leukemogenesis and in
As mentioned before, after chemotherapy, rem- discovery of molecular markers for BCP-ALL
nants of blast cells are one of the most important prog- diagnosis, prognosis and patient response to
nostic factors for BCP-ALL patients [34–36]. While FC treatment
has become an important tool for the detection of posi- MS is a high-throughput technology has been used for
tive MRD, the modulation of surface antigens during quantitation and identification of unknown molecules
therapy of B-ALL can affect the results [37]. For and for the investigation of different pathologies
example, analysis by FC of BM from children with such as rheumatoid arthritis, diabetes, heart diseases
BCP-ALL at 15 and 33 days of treatment showed a and different cancers, including leukemia [42,43]. MS
downregulation of the surface markers CD10, CD19 commonly involves the digestion, ionization, and
and CD34, and increased expression of CD20; capture of the proteins by the MS system and later,
however, no changes in the expression of CD38, the detection of the molecules according to the
CD58 and CD45 was reported [38]. To overcome this mass-to-charge ratio (m/z) [44]. Mass spectrometers
challenge, Theunissen et al., performed the standardiz- consist of three basic components: an ion source, a
ation of different markers to improve the specificity mass analyser, and an ion detector. MS measurements
and sensitivity of MRD diagnostic based on FC. They are carried out on ionized analytes in the gaseous
standardized two different sets of antibodies to analy- phase, requiring a method to transfer molecules from
sis different BM samples derived from pretreated and solution or solid phase into this state. The two most
relapse patients with BCP-ALL. The use of the anti- commonly used techniques are matrix-assisted laser
bodies CD19, CD10, CD20, CD34 and CD45 in combi- desorption/ionization (MALDI) and electrospray ioniz-
nation with CD38, CD81 and CD15/NG2, could ation (ESI) [45,46]. Both MALDI and ESI are soft ioniz-
discriminate normal and leukemic cells in 99% of ana- ation techniques in which ions are created with low
lyzed samples [39]. Furthermore, FC showed a high internal energies and thus undergo little fragmenta-
degree concordance of 98% in comparison to the tion. Electrospray and matrix-assisted laser desorption
specificity of Polymerase Chain Reaction (PCR)-based ionization processes provide ‘soft’ (meaning low-
MRD detection [39,40]. In a similar manner, the energy) ionization methods for a variety of
HEMATOLOGY 641

Table 2. Examples of potential molecular biomarkers for diagnosis, prognosis, and targeted therapy of BCP-ALL.
Biomarkers Functions Applications in B-ALL References
CD79a B-cell activation. Diagnosis of B-ALL and subtype classification. [29,35–38]
CD38 Catalyzes the synthesis from cADP ribose to ADP ribose. It is used for B-ALL diagnosis. [30–32]
TP53, JAK2 Tumor suppressor-transcription factor, Participates in intracellular Both molecules have been tested as potential [101–103]
signaling upon activation of cytokine receptors. biomarker of poor outcome in B-ALL. [39–41]
RAS GTPase involved in protein cell signaling. Unfavorable prognosis and predictive [104,105]
biomarker for treatment. [42,43]
LRG1, CLU, F2, Participates in vascularization by modulating TGF-β signaling, This cluster of proteins have the potential to [52,106,107]
SERPIND1 Promotes or inhibits tumorigenesis based in the cellular context, be used for B-ALL diagnosis and treatment [44–46]
Anticoagulant and proinflammatory, Coagulating process. evaluation.
CSRP2 Participates in breast cancer cell invasiveness. Diagnosis/treatment monitoring for B-ALL. [47–108]
ADA1 Implicated in purine salvage pathway. Diagnosis/treatment monitoring for B-ALL. [48–109]
IKZF1, CRLF2 Participates in the regulation of hematopoiesis, Proto-oncogene in Both molecules can be used as markers for [49,50,110,111]
B-ALL. poor prognosis in high-risk ALL.
Hsp90 Participates for folding and regulation of cellular proteins. Diagnosis/treatment monitoring for B-ALL. [51,52,112,113]
A20 Inhibits activation of NF-κβ and apoptosis. Target for B-ALL treatment. [53,114]

biomolecules, including peptides, proteins, drug abrogates the IK6 phenotype by promoting expression
metabolites, oligonucleotides and carbohydrates, to of IKZF1, demonstrating the role of IKZF1 alteration in
enable their measurement by MS [47]. the leukemogenesis process and its potential thera-
There are several mass spectrometers that combine peutic use for BCP-ALL. In another study, Saha et al.,
ESI or MALDI with a variety of analysers. In MALDI, by using 2D-PAGE followed by matrix-assisted laser
samples are co-crystallised with an organic matrix on desorption/ionization time of flight-tandem MS
a metal target. A pulsed laser is used to excite the (MALDI-ToF-MS/MS), could identify 79 proteins differ-
matrix, which causes rapid thermal heating of the mol- entially regulated in CD19+ cells from BCP-ALL patients
ecules and eventually desorption of ions into the gas [51]. Ontology analysis of these proteins demonstrated
phase. Because of the usage of a pulsed laser, MALDI their participation in proteostasis, cytoskeletal organiz-
produces packets of ions rather than a continuous ation, and signal transduction, thus suggesting their
beam. This ionization technique tolerates a reasonable potential implications in BCP-ALL leukemogenesis.
amount of impurities in the sample to be analyzed. On When BCP-ALL has already been established, early
the other hand, ESI is based on spraying an electrically diagnosis is important for better management and to
generated fine mist of ions into the inlet of a mass significantly contribute in the primary induction
spectrometer at atmospheric pressure. This technique therapy for the remission of the disease. Cavalcante
ionizes molecules directly from solution, so it can et al., by using lectin affinity chromatography and
easily be interfaced with liquid separation methods liquid chromatography-MS (LC-MS) identified 96
[48]. After ionization, the sample reaches the mass ana- different proteins in serum samples of patients diag-
lyser, which separates ions by their mass-to-charge nosed with BCP-ALL [52]. Leucine-rich alpha-2-glyco-
ratios. Ion motion in the mass analyser can be manipu- protein 1, Clusterin, Thrombin, Heparin Cofactor II,
lated by electric or magnetic fields to direct ions to a Alpha-2-macroglobulin, Alpha-2-antiplasmin, Alpha-1
detector, which registers the numbers of ions at each antitrypsin, and Complement factor B and C3 were
individual m/z value. over expressed in BCP-ALL in comparison with the
In proteomics research, four basic kinds of mass ana- induction therapy group, which means that candidate
lysers are currently being used: time-of-flight (ToF), ion biomarkers only are seen in the disease state and are
trap, quadrupole (single and triple or tandem), and therefore proposed as potential biomarkers for diagno-
Fourier transform ion cyclotron resonance (FTICR) ana- sis and favorable response after induction therapy [52].
lysers. All four differ considerably in sensitivity, resol- In a similar manner, Mirkowska et al., by using a combi-
ution, mass accuracy and the possibility to fragment nation of LC-MS and FC, analyzed the surface protein
peptide ions [44]. The combination of an ion source, composition of B-cell precursors of BCP-ALL patients
mass analyser and detector is usually determined by xenografted on immunodeficient mice [53]. They ident-
the application. The human blood proteome is fre- ified 713 surface proteins, including glycosylated and
quently assessed by protein abundance profiling lysine-containing proteins (hematopoietic and B-cell
using tandem mass spectrometry [49]. markers). FC validation of some of these molecules
The use of tandem-MS has demonstrated that Phila- (CD63, CD97, CD157, and CD305) in human samples
delphia chromosome positive (Ph+) B cell expressing highlighted their potential uses in the diagnosis of
IK6 showed increased expression of a set of adhesion BCP-ALL.
molecules, including thymocyte differentiation Another example of the use of MS in the identifi-
antigen 1 (THY1, CD90), L-selectin, and THY1 ligand cation of new molecules for diagnosis and risk stratifi-
integrin alpha 5, leading to an aggressive lymphoid leu- cation in BCP-ALL is the study carried out by Xu et al.
kemia phenotype [50]. Interestingly, retinoid treatment [54]. Authors analyzed by liquid chromatography-
642 A. F. CITALAN-MADRID ET AL.

Table 3. Proteomic tools in studies of BCP-ALL.


Type of
Tool study Objective Findings References
LC-ESI-MS In vitro To analyze the differential protein expression in the Vincristine treatment led to the expression of 135 [83]
pediatric B-cell line CCRF-SB during vincristine proteins participating in Toll receptor signaling
treatment. pathway, Ras Pathway, B and T cell activation, CCKR
signaling map, cytokine-mediated signaling
pathway, and oxidative phosphorylation.
MFC Case- To analyze the expression of B-, myeloid, T-lymphoid Aberrant expression of myeloid-associated antigens [33]
control and NK-cell antigens in 200 blood or bone marrow was observed in 86.5% of cases and differential
samples of pretreated children and adult patients expression between children and adult patients
with B-ALL. with B-ALL.
MFC Case- To analyze the differential expression of TdT, CD34, Differential expression was observed between CD10+ [32]
control CD45, CD10, CD38, CD20, and CD22 in CD10+ and in CD10- subpopulations. CD10+ blasts exhibited
Cd10- subpopulations of B-ALL cells and higher levels of TdT, CD22, CD34, and CD20.
hematogones. Conversely, CD10- blasts showed higher expression
of CD45 than CD10+ blasts.
MFC Case- To identify the malignant clone harboring clone-specific FC could identify 93% true-positive (leukemic) and [115]
control genomic markers in 53 bone marrow samples derived 93% true-negative (normal) cell populations and
from 28 children diagnosed with B-ALL for B-ALL MRD four discrepant cases for MRD monitoring in B-ALL
monitoring. cases.
MFC Case- To analyze the immunomodulation of cell surface- A downregulation of CD10, CD19, and CD34 and [38]
control antigen in MRD-positive samples of children with B- upregulation of CD20 were observed after 15 and
ALL for MRD detection. 33 days of treatment, but no changes in the
expression of CD38, CD58 and CD45 were noticed.
MFC Case- To standardize an immunophenotyping protocol for A standardized FC protocol showed a high percentage [39]
control MRD monitoring and for sensitivity improvement in of concordance (93%) in comparison with RQ-PCR-
319 bone marrow samples of children with B-ALL based MRD detection.
after induction therapy.
MFC Case- To standardize a multicolor FC protocol to evaluate the FC analysis by using a set of B-cell markers showed a [40]
control MRD in 263 bone marrow samples derived from consistently high overall concordance (P < .001)
childhood relapsed B-ALL. and, under optimal conditions, sensitivity
comparable to the PCR method for MRD detection.
MFC Case- To analyze the expression of CD73 in normal young CD73 expression was 6-fold greater in MRD-positive B [116]
control hematogones cells and B-ALL cells with or without cells in comparison with MRD negative ones. In all,
MRD+ of bone marrow for MRD monitoring. 41.82% MRD-positive B-ALL cases expressed high
CD73 and sensitivity MRD detection reached 10−4.
MFC Case- To analyze the expression of Neurophilin-1 (NRP-1)/ CD304 expression was higher in 40% of B-ALL (CD304 [41]
control CD304 as an MRD and prognosis marker in bone +) cells at diagnosis compared with controls.
marrow samples derived from 70 children with B-ALL However, at day 28 of induction therapy, 57.1% of
and 40 controls. CD304+ patients transitioned to a CD304-
phenotype.
LC-MS/MS Case- To analyze protein expression for risk stratification in 86 differently expressed proteins were identified in [54]
control BM samples derived from B-ALL patients. The patients childhood high-risk ALL patients. They found the
were classified into low /medium and high-risk aberrant evens happened in pre-mRNA splicing,
groups. DNA damage response, and stress response.
AC-MS Case- To analyze the protein expression profile and biomarker A panel of molecules (i.e. LRG1, CLU, F2, SERPIND1, [52]
control discovery in 10 samples derived from B-ALL patients. A2M, SERPINF2, SERPINA1, CFB, and C3) were
identified as potential biomarkers for early
diagnosis of B-ALL.
LC-MS/FC In situ/in To analyze the protein expression profile in A total of 713 proteins were detected in B-ALL cases. [53]
vivo immunodeficient mice xenotransplanted with B-ALL From all these proteins, CD97, CD157, CD63, and
human cells. CD305 were identified as the most promising
makers to distinguish between normal and
malignant cells.
SELDI-ToF- Case- To analyze the protein expression profile and biomarker PF4, CTAP-III, and two fragments of C3a were [117]
MS control discovery in samples derived from ALL pediatric differentially expressed and used to distinguish
patients. pediatric BCP-ALL patients from healthy controls,
and pediatric AML patients.
LC-MS/MS Case- To analyze the role of IKZF1 in B-ALL leukemogenesis. Ikzf1 and Arf alterations contribute to leukemogenesis [50]
control by promoting the development of an aggressive
lymphoid leukemia.
2D-PAGE/ Case- To analyze protein expression profiles and protein A total of 79 differentially regulated proteins were [51]
MALDI- control functions in B-ALL patient samples. identified in B-ALL cells, participating in
ToF-MS/ proteostasis, cytoskeletal organization, redox
MS homeostasis, and signal transduction pathways
relevant to leukemogenesis.
SDS-PAGE/ Case- To analyze the expression and function of Aurora B Expression of AURKB and AURKA was mostly [69]
WB/RPPA control kinases in 172 B-ALL pediatric patients. observed in E2A-PBX1-translocated B-ALL cases.
AURKB inhibition resulted in proliferation arrest and
apoptosis.
RPPA Case- To analyze the protein expression profiles for Increased phosphorylation of ErbB2 was observed in [70]
control/ identification of new therapeutic targets in 129 56% of Ph+ ALL. ErbB2 kinase activity inhibition by
in vitro samples derived from Ph+ B-ALL patients and by canertinib resulted in increased expression of the
using Z-181 and Z-119 cell lines. proapoptotic protein Bim, caspase-3 activation, and
cell death.
RPPA Case- To analyze the expression of cathepsin G (CG) in 130 CG was overexpressed in B-ALL samples and was [72]
control samples derived from B-ALL patients. proposed as a poor prognostic marker. Endogenous
expression analysis demonstrated that CG can also

(Continued)
HEMATOLOGY 643

Table 3. Continued.
Type of
Tool study Objective Findings References
be taken up by different B-ALL cell lines (SB, RS4,
Nalm6, and HMY).
RPPA Case- To analyze the role of the STAT5 signaling pathway in Increased phosphorylation of STAT5, which was [73]
control/ leukemogenesis by using samples derived from B-ALL correlated with poor prognosis in BCR-ABL+ ALL
in vivo patients and the Stat-CA mouse model. patients.
RPPA Case- To analyze the role of PAX5-JAK2 fusion protein in B- It was observed that the PAX5-JAK2 fusion protein led [74]
control ALL to phosphorylation of STAT1, 3, and 5. Additionally
JACK2 inhibition led to the death of transformed
cells.
RPPA In vitro/in To analyze the role of the Wnt signaling pathway in Marrow stromal cells (MSC) induced activation of the [81]
vivo leukemogenesis by using the Reh cell line and a Wnt signaling pathway of leukemic cells to induce
mouse leukemia model. drug resistance and cell survival.
RPPA Case To analyze the role of the PI3K-Akt-mTOR signaling It was demonstrated that reduced expression of the [82]
control/ pathway in leukemogenesis by using 72 samples CD200 protein was associated with increased
in vitro derived from B-ALL patients and the Reh cell line. activation of the PI3K-Akt-mTOR signaling pathway
in B-ALL patients, characterized by increased
phosphorylation of Akt.
RPPA Case- To analyze the role of TCF3-PBX1 fusion in cell TCF3 + B-ALL samples showed significant expression [75]
control/ proliferation and differentiation in samples derived of elements of the pre-BCR signaling complex
in vitro from 19 TCF3-rearranged and 113 non-TCF3 B-ALL (ZAP70, SLP65, BLNK, BTK, PI3K-p110δ, and IRF4).
pediatric patients and by using the MHH-CALL3 cell BTK inhibition led to decreased phosphorylation of
line. Erk1/2 and reduction of growth and cell viability.
RPPA In vitro To analyze the role of hypoxia in leukemic cell survival Hypoxia induced chemoresistance by increasing the [80]
chemoresistance to methotrexate (MTX) and expression of anti-apoptotic proteins (XIAP, Mcl-1,
prednisolone (PRD) in the Nalm-6 and Reh B-ALL cell and Bcl-2) and decreasing the expression of pro-
lines. apoptotic proteins (cleaved caspase 3, Bax, and
Bim).
RPPA Case- To analyze the role of the Erk1/2 signaling pathway in Increased expression of phospho-ERK-T202/Y204, [76]
control/ leukemogenesis by using 192 samples derived from DUSP6, SPRY2, and ETV5 was observed in CD19+
in vitro B-ALL patients and leukemic pre-B cells. bone marrow pre-B cells and patient-derived pre-B
ALL. DUSP6 inhibition produced hyperactivation of
Erk but increased ROS levels and cell death.
CyTOF Cases To identify characteristics of expanded leukemic Were identified 6 features of expanded leukemic [118]
populations sufficient to predict relapse at diagnosis populations sufficient to predict patient relapse at
of BCP-ALL. diagnosis. These features implicated pro-BII cells
with activated mTOR signaling, and pre-BI cells with
activated and unresponsive pre-B-cell receptor
signaling.
CyTOF Cases/in To identify additional activated networks in BCP-ALL SCR/ABL inhibition as effective in disrupting the [119]
vitro primary patient samples by single-cell mass cooperative functional networks present in hiCRLF2
cytometry. BCP-ALL patients, supporting further investigation
of this strategy in pre-clinical studies.
CyTOF Case- To analyze the depth of single-cell mass cytometry and Single-cell trajectory detection uncovers progression [120]
control an algorithm developed to predict the developmental and regulatory coordination in human B cell
path de novo of human B cell lymphopoiesis. development. This trajectory revealed nascent
fractions of B cell progenitors and aligned them
with developmentally-cued, regulatory signaling
including IL-7/STAT5.
Note: Proteomics tools such as FC, 2D-PAGE, MS, and RPPA have aroused great interest in research and the discovery of new biomarkers for the diagnosis,
prognosis, and monitoring of MRD and for the identification of new therapeutic targets for BCP-ALL.

tandem MS (LC–MS/MS) the protein expression profile the last years, mass cytometry (CyTOF) emerged as a
in 12 BM samples of BCP-ALL from patients classified new technology for high-dimensional multi-parameter
as low/medium and high-risk groups, and 6 patients single cell analysis that overcomes the limitations of
with non-malignant hematological disorders (non- conventional flow cytometry [56]. Recently there has
ALL group). Considering the non-ALL group as refer- been rapid progress in multiparametric single cell pro-
ence, authors identified 86 differentially expressed teomic and genomic research. CyTOF is a single cell
proteins in the high-risk childhood B-ALL group, proteomics platform that combines elemental MS
including Elongation factor 1-alpha 1, Thymosin with FC enabling up to 50 parameters per single cell
alpha-1 and Histone H3.1. Interestingly, a total of 35 to be measured [57], it has become a revolutionary
of these proteins were predicted to have directive tool to investigate the hematopoietic system [57,58].
protein–protein interactions and participate in pre- The high number of parameters facilitates analysis of
mRNA splicing, DNA damage response and stress highly complex cell populations and could be extre-
response which could explain the high-risk outcome mely useful for the diagnosis of malignant hematologic
for BCP-ALL. disorders, monitoring of MRD, and selection of treat-
ment modalities [59]. In CyTOF, the fluorophore-
2.2.1. Mass cytometry (CyTOF) labeled antibodies are replaced by antibodies of isoto-
Over time, cytometry methods have been a powerful pically purified heavy metal. The presence of the bound
tool to investigate the hematopoietic system [55]. In antibodies is detected through the use of inductively
644 A. F. CITALAN-MADRID ET AL.

coupled plasma ionization and time-of-flight mass tool to achieve this goal. An example is a study
spectrometry (ICP-MS TOF) analysis of the metal ions carried out by Hartsink-Segers et al. who analyzed the
that were attached to each antibody. The advantage expression of Aurora A (AURKA) and Aurora B kinases
of this approach stems from the ability of ICP-MS to dis- (AURKB) by RPPA in combination with SDS-PAGE and
tinguish ions of different atomic weight with less than western blot [69]. Expression of both kinases was posi-
1% signal spillover between adjacent masses [59]. tive in 172 samples derived from BM or PB of pediatric
CyTOF allows for the measurement of over 40 samples BCP-ALL. Even more, in vitro downregulation
markers simultaneously without the concern of spec- of AURKB showed anti-proliferative and pro-apoptotic
trum overlap. It has enabled the analysis of intact biologi- effects, suggesting that AURKB can play an important
cal specimen without pre-depletion steps or a dump role in B-ALL leukemogenesis and demonstrating its
channel. Therefore, we can view the hematopoietic utility as a possible target for leukemia treatment.
system comprehensively with high-content dimension- Like AURKB, the human epidermal growth factor recep-
ality from conventional 2-D flow cytometry plots [55]. tor 2 (ErbB2) kinase has been shown to play an impor-
The high number of parameters afforded by mass cyto- tant role in the BCP-ALL pathological process. Irwin
metry enables the organization of numerous distinct et al. studied 129 samples derived from Ph+ BCP-ALL
cell populations within very complex cell samples, patients by using an RPPA methodology [70] and
while still allowing sufficient additional measurement they found overexpression of ErbB2 in 56% of these
parameters to characterize functional properties of the patients. In vitro inhibition of ErbB kinase activity in
different cell populations [59]. High-parameter analysis the B-cell leukemic cell lines Z-181 and Z-119 led to a
of malignant diseases, particularly leukemias, is an area decreased proliferation and increased sensitivity to
in which mass cytometry could be particularly useful in BRC/ABL TKI treatment. Thus, the ErbB signaling
the clinic, and initial experiments have been performed pathway could be an attractive pathway for new thera-
by several research groups [60–64]. These studies high- peutic interventions in Ph+ BCP-ALL.
light a particular strength of high-parameter analysis: Another potential target for BCP-ALL is cathepsin G
the ability to define rare cell types, such as leukemia (CG). CG is normally expressed in AML [71], however, its
stem cells, and simultaneously characterize the func- expression in BCP-ALL has been studied by Khan et al.
tional properties of these cells. Table 3 shows additional [72]. By performing an RPPA analysis of 130 patients
studies that complement the applications of CyTOF in with BCP-ALL, they showed increased expression of
the diagnosis and prognosis of BCP-ALL. CG in a high proportion of the samples; therefore, CG
can be a potential target for therapy in BCP-ALL. On
the same sense, to unveil the role of signal transducer
2.3. Protein arrays for the study and
and activator of transcription 5 (STAT5) in BCP-ALL, Hel-
identification of therapeutic targets of BCP-ALL
temes-Harris et al., by using an RPPA methodology, per-
Protein array is a high-throughput technology based formed an analysis of 129 samples collected from the
on a set of antigens or antibodies arranged onto solid blood and/or BM of BCP-ALL patients [73]. The
support for protein identification and functional analy- expression of STAT5 was similar in all analyzed
sis. There are three different configurations for protein samples, but increased phosphorylation of this mol-
arrays identified as analytical, functional and reversed ecule was observed in a subset of Ph+ BCP-ALL
phase protein array (RPPA) [65]. Unlike analytical and patients. Similarly, Schinnerl et al. [74] analyzed by
functional arrays, RPPA works by immobilizing the RPPA a set of PAX5-JAK2+ BCP-ALL samples from BM
interest samples (e.g. tissue, cell lysates or fractionated patients and found increased phosphorylation of
cell lysates) onto a solid surface for protein detection STAT5; thus, the fusion protein PAX5-JAK2 could act
by using a set of antibodies [66]. It is possible to as an upstream activator of STAT5, which promotes leu-
screen thousands of interactions in parallel – protein- kemogenesis, and could be used as a therapeutic
antibody, protein–protein, protein-drug, enzyme–sub- target for BCP-ALL.
strate – as well as providing potential assays for bio- Pre-B cell receptor (pre-BCR) is expressed on the
markers. The different types of arrays have surface of pre-B cell precursors, and its activation has
applications in clinical diagnostics, which includes an important role in the clonal propagation of pre-B
detection of antibodies in autoimmune diseases or bio- cells in BM. Thus, alterations in pre-BCR signaling
markers in sera of cancer patients, as well as in target could contribute in the leukemogenesis process. At
and drug discovery [67,68] (Figure 2). this respect, van der Veer et al., by using an RPPA meth-
odology demonstrated that pre-BCR signaling is
2.3.1. Reversed phase protein array usefulness in affected by TCF3 gene rearrangement in BCP-ALL
the search of biological targets of BCP-ALL [75]. From 19 TCF3-rearranged and 113 non-TCF3
The identification of biological targets with significance BCP-ALL pediatric samples analyzed by RPPA, the
in BCP-ALL is very useful for the development of new TCF3-rearranged group showed more significant
therapeutic approaches of disease. RPPA is a powerful expression of various elements of the pre-BCR
HEMATOLOGY 645

Figure 2. A protein microarray (protein chip) is a high throughput method used to track the interactions and activities of proteins.
Protein microarrays are typically prepared by immobilizing proteins onto a microscope slide using a standard contact spotter or
non-contact microarray. There are three types of protein microarrays that are currently used to study the biochemical activities
of proteins: Analytical protein microarrays are mostly represented by antibody arrays and focus on protein detection. In this
class of microarrays, targeted proteins can be detected either by direct labeling or using a reporter antibody in sandwich assay
format. Functional protein microarrays have broad applications in studying protein interactions, including protein binding and
enzyme-substrate reactions. Reverse-phase protein microarrays provide a different array format by immobilizing many different
lysate samples on the same chip. There are five major areas where protein arrays are being applied: diagnostics, proteomics,
protein functional analysis, antibody characterization and treatment.

signaling, including ZAP70, SLP65, Bruton’s tyrosine DUSP6 produced the hyperactivation of Erk, the incre-
kinase (BTK), PI3K-p110δ, and IRF4. Corresponding to ment of ROS levels and cell death, suggesting that the
its role as an effector of BTK, in vitro analysis in MHH- Erk signaling pathway is controlled by negative feed-
CALL3 cells showed decreased phosphorylation of back through SPRY2, ETV5, and DUSP6, and it can rep-
Erk1/2 and growth and cell viability after treatment of resent an interesting target for BCP-ALL treatment.
BTK inhibitor Ibrutinib. Therefore, interference with
pre-BCR signaling is an excellent candidate for a thera-
2.4. RPPA and MS as approaches useful in the
peutic target for TCF3-rearranged BCP-ALL. In a similar
understanding of the molecular basis of
study, the proteomic profile of blood and BM from 192
treatment resistance in BCP-ALL
BCP-ALL patients was analyzed by Shojaee et al. [76]. By
mean an RPPA methodology, they observed an Leukemic cells can promote mechanisms that enable
increased expression of phospho-ERK-T202/Y204, the development of resistance to leukemia anticancer
DUSP6, SPRY2 and ETV5 in normal CD19+ BM pre-B drugs, thereby leading to relapse and a poor
cells and BCP-ALL. In vitro, overexpression of DUSP6 outcome of the disease [77,78]. Thus, the search for
led to pre-B cell transformation and colony formation. strategies against leukemia drug resistance to
Conversely, the 2-benzylidene-3-(cyclohexylamino)-1- develop new therapies have been recently revised
indanone hydrochloride (BCI)-induced inhibition of [79]. Hypoxia is one the mechanism used for leukemic
646 A. F. CITALAN-MADRID ET AL.

cells to promote cell survival and drug resistance that, in the best of cases they could be the object of
during BCP-ALL treatment. The analysis of protein targeted therapies.
expression of Nalm-6 and Reh B-ALL cells under nor-
moxic conditions by RPPA showed that the treatment
3. Validation of biomarkers as a pre-
of methotrexate (MTX) and prednisolone (PRD) lead
requisite for its clinical application
to a decreased cell viability Petit et al. [80]. However,
hypoxic conditions lead to overregulation of proteins MS, RPPA, 2D-PAGE and/or WB have aroused great
related to drug resistance and deregulation of pro- interest in research and the discovery of new bio-
apoptotic proteins accompanied by the expression of markers for diagnosis, prognosis, monitoring of MRD
the anti-apoptotic proteins XIAP, Mcl-1, and Bcl-2. Fur- and for the identification of biological targets for
thermore, cell viability was higher, suggesting a protec- BCP-ALL. However, before proceeding with any clinical
tive role of hypoxia and anti-apoptotic signals in application, the confirmation of the usefulness of
leukemogenesis, regulating cell death and promoting potential protein markers must be assessed in a pro-
cell survival. spective or retrospective manner [84]. Once the
The regulation of the BM microenvironment marker has been approved for clinical practice, its
through the Wnt signaling pathway is another factor detection and measurement must be rapid and easy
intervening in drug resistance affecting the outcome to perform, considering quality parameters such as
of BCP-ALL [81]. As demonstrated by Yang et al., by maximum sensitivity and specificity. Unfortunately,
mean of RPPA, the analysis of protein expression such requirements are not entirely filled by the most
under treatment of cytarabine (Ara C) lead to an of the molecular markers reported, and the search for
increased expression of 154 proteins related to antia- new biomarkers is an arduous task, because it involves
poptotic proteins, drug resistance and different the collection of several samples and then, the analysis
members of the Wnt signaling pathway, such as Lef1, of thousands of potential molecules (Table 3).
c-Myc and CCNDBP1. Furthermore, in vivo blocking of In addition, determination of the biomarker needs
Wnt signaling sensitizes leukemia cells to drug treat- to be performed in a readily obtainable specimen
ment and improved overall survival, demonstrating (e.g. blood, serum, urine, etc.) and monitored by a
its potential therapeutic target. Another studied mech- non-invasive method. It is desirable that the method
anism is mTOR. By mean of activation of the Wnt is performed on as many samples as possible by
pathway, mTORC1 regulates the cell cycle and affects using accessible equipment. Consequently, assessment
the leukemogenesis process. At this respect, Ghazavi and interpretation of molecular biomarkers are com-
et al., by using an RPPA approach analyzed 72 monly performed through a series of molecular,
samples derived from ETV6/RUNX1+ BCP-ALL patients immunological, or proteomic tools that must be vali-
[82]. They found that many of these patients showed dated for their dynamic range of detection and repro-
decreased expression of the glycoprotein CD200 ducibility [85].
which corresponded to increased expression of PI3 K, As for diagnosis and leukemia classification, the
Akt kinases and mTOR. Thus, it seems to be possible identification and validation of biomarkers for BCP-
that, the use of inhibitors for these kinases could rep- ALL MRD monitoring have been performed routinely
resent a viable approach for ETV6/RUNX1+ BCP-ALL by FC. An example of one study in which both the
treatment. identification and validation of protein markers were
Vincristine, a vinca alkaloid, interacts with tubulin carried out, was that performed by Tembhare-Prashant
disrupting microtubule polymerization and favoring et al. In their study, authors analyzed the expression of
the cell cycle arrest in the M phase, (which is fol- CD surface molecules for MRD monitoring in 90 chil-
lowed by induction of apoptosis), is used in dren with B-ALL [86], observed a differential expression
several stages of the ALL treatment [83]. In vitro of CD24, CD44, CD72, CD73, CD86 and CD200 between
studies performed by Guzmán-Ortiz et al., showed hematogones and B-ALL cells after 35 days of induction
a differential protein expression in the pediatric B- therapy. In MRD-positive samples, CD73 showed the
cell line CCRF-SB during gradual exposition to vin- maximum (83%) frequency of leukemia- associated
cristine [83]. Complementary Liquid Chromato- immunophenotype (LAIP) and CD86, the highest
graphy-Electro Spray Ionization-MS (LC-ESI-MS) (100%) stability of aberrant expression. Sherif et al.,
analysis, demonstrated that vincristine treatment observed that after 14 days of post-induction treat-
induced the expression of 135 proteins related ment, there was a reduction in the expression of
with Toll receptor signaling, Ras pathway, B and T surface markers CD10, CD19, CD34 and CD97 in 30
cell activation, CCKR signaling, cytokine-mediated pediatric patients diagnosed with BCP-ALL [87]. Inter-
signaling pathway, and oxidative phosphorylation. estingly, at day 14, in 81.5% of these patients, CD97
In this way, it is logical to think that each of these was undetectable. Therefore, the use of these surface
mechanisms would be contributing to the resistance markers for MRD monitoring in children with BCP-ALL
to pharmacological treatment by leukemic cells and should be considered.
HEMATOLOGY 647

4. Concluding remarks and challenges genomics or metabolomics, which must comply with
standardized and valid guidelines.
Proteomics has been successfully used to measure the
The translation of a potential biomarker from basic
expression of proteins in an organism using samples
investigation to clinical practice involves a slow and
from different biological compartments. However, the
gradual process, which depends mostly on the qualifi-
complexity of biological samples increased the possi-
cation of the tool and the validation of the biomarker
bility of erroneous analysis of molecules. Furthermore,
itself. Some of these reasons mean that the identifi-
there are not only limitations regarding the perform-
cation of potential biomarkers by protein detection-
ance or applicability of proteomics tools in the investi-
based tools does not yet have the expected impact
gation of BCP-ALL, but there are also several drawbacks
on decision making within clinical practice. However,
that have their origins in sample or manipulation
the incorporation of bioinformatics and protein data-
during its processing, which can be translated into
bases have facilitated the analysis of information pro-
the loss of potential biomarkers for diagnostic or thera-
duced by high-throughput proteomic method
peutic purposes.
expanding the use of proteomic tools beyond basic
Since BCP-ALL research based on proteomics tools
research. In the near future, proteomic tools such as
requires the correct sampling of either the cytosolic
MS, CyTOF, RPPA or FC, could yield better performance
or membrane proteins, this will require an adequate
as long as established protocols consider, for instance,
extraction method and a suitable reagent that does
sample collection and storage, the well-controlled
not interfere with the physicochemical properties of
design of case-control studies, and the reduction of
the molecule (i.e. solvent, lysis buffer) [88]. Further-
costs in their applications. However, it is undeniable
more, approaches aimed at the search for potential
that applications of these tools have increased our
biomarkers derived from blood samples become
knowledge of BCP-ALL, and they have had a positive
more complicated, due to the complexity and the enor-
impact on the diagnosis and treatment of this disease.
mous abundance of proteins contained in human
serum. In this way, one of the main challenges is the
elimination of serum albumin, which represents up to Disclosure statement
40% of the total protein in the intravascular space No potential conflict of interest was reported by the authors.
[89]. Even when available methods to eliminate, this
and other abundant proteins are efficient, the loss of
Funding
those proteins with less abundance and that represent
potential candidates as biomarkers remain as the main This work was funded in part by the following National
challenge. Council of Science and Technology (Consejo Nacional de
Ciencia y Tecnología, CONACYT) grant numbers [-SALUD-
On the other hand, collection of the specimen being
2016-01-273144, -INF-2014-225520, and -INFR-2015-01-
analyzed is of vital importance, and this must be done 254106].
in the most efficient manner and in the shortest time.
Whether the sample is derived from a specific tissue
or from blood, these should be collected and following ORCID
a standardized protocol minimizing errors to obtain Marisol Ibarra-Ramirez http://orcid.org/0000-0002-3875-
real and representative results. For example, some of 4532
the variables that may affect the results obtained by Idalia Garza-Veloz http://orcid.org/0000-0002-6307-1696
Margarita L. Martinez-Fierro http://orcid.org/0000-0003-
blood samples may include the type of additive or 1478-9068
anticoagulant used (i.e. heparin, EDTA or sodium
citrate), temperature and processing time, hemolysis
of the sample, storage temperature of the sample, as References
well as the processes of freezing and thawing the [1] Fiegl, M., Epidemiology, pathogenesis, and etiology of
sample. acute leukemia; 2016. p. 3–13.
Although proteomics offers enormous advantages [2] Loghavi S, Kutok JL, Jorgensen JL. B-acute lymphoblas-
through the analysis of large amounts of sample, tech- tic leukemia/lymphoblastic lymphoma. Am J Clin
Pathol. 2015;144(3):393–410.
nical obstacles still do not allow its basic or clinical
[3] Zhang X, et al. B lymphoblastic leukemia/lymphoma:
application in a routine way. Moreover, the costs in new insights into genetics, molecular aberrations, sub-
the proteomic tools and their complexity, the high classification and targeted therapy. Oncotarget. 2017;8
prices in the consumables as well as trained personnel, (39):66728–66741.
represent a real inconvenience at the time of their [4] Siegel DA, et al. Rates and Trends of pediatric acute
application. On the other hand, we must point out lymphoblastic leukemia – United States, 2001-2014.
MMWR Morb Mortal Wkly Rep. 2017;66(36):950–954.
the enormous challenge represented by the inte- [5] Yilmaz M, Kantarjian H, Jabbour E. Treatment of acute
gration of data derived from proteomics research, lymphoblastic leukemia in older adults: now and the
with those obtained in other branches, such as future. Clin Adv Hematol Oncol. 2017;15(4):266–274.
648 A. F. CITALAN-MADRID ET AL.

[6] Perez-Saldivar ML, et al. Childhood acute leukemias are [27] van Zelm MC, et al. Ig gene rearrangement steps are
frequent in Mexico City: descriptive epidemiology. initiated in early human precursor B cell subsets and
BMC Cancer. 2011;11:355. correlate with specific transcription factor expression.
[7] Bernaldez-Rios R, et al. The age incidence of childhood J Immunol. 2005;175(9):5912–5922.
B-cell precursor acute lymphoblastic leukemia in [28] Campo E, et al. The 2008 WHO classification of lym-
Mexico City. J Pediatr Hematol Oncol. 2008;30(3):199– phoid neoplasms and beyond: evolving concepts
203. and practical applications. Blood. 2011;117
[8] Tijchon E, et al. B-lineage transcription factors and (19):5019–5032.
cooperating gene lesions required for leukemia devel- [29] Jiang Z, et al. CD34 and CD38 are prognostic bio-
opment. Leukemia. 2013;27(3):541–552. markers for acute B lymphoblastic leukemia. Biomark
[9] Garza-Veloz I, et al. Identification of differentially Res. 2016;4:23.
expressed genes associated with prognosis of B [30] Coustan-Smith E, et al. A simplified flow cytometric
acute lymphoblastic leukemia. Dis Markers. assay identifies children with acute lymphoblastic leu-
2015;2015:828145. kemia who have a superior clinical outcome. Blood.
[10] Quezada H, et al. Omics-based biomarkers: current 2006;108(1):97–102.
status and potential use in the clinic. Bol Med Hosp [31] Bene MC, et al. Immunophenotyping of acute leukemia
Infant Mex. 2017;74(3):219–226. and lymphoproliferative disorders: a consensus propo-
[11] Foss EJ, et al. Proteomic classification of acute leuke- sal of the European LeukemiaNet work Package 10.
mias by Alignment-based quantitation of LC–MS/MS Leukemia. 2011;25(4):567–574.
data sets. J Proteome Res. 2012;11(10):5005–5010. [32] Sedek L, et al. The immunophenotypes of blast cells in
[12] Chandramouli K, Qian PY. Proteomics: challenges, tech- B-cell precursor acute lymphoblastic leukemia: how
niques and possibilities to overcome biological sample different are they from their normal counterparts?
complexity. Hum Genomics Proteomics. 2009;2009:1– Cytometry B Clin Cytom. 2014;86(5):329–339.
22. [33] Seegmiller AC, et al. Characterization of immunophe-
[13] Srinivas PR, et al. Proteomics for cancer biomarker dis- notypic aberrancies in 200 cases of B acute lympho-
covery. Clin Chem. 2002;48(8):1160–1169. blastic leukemia. Am J Clin Pathol. 2009;132(6):940–
[14] Hudler P, Kocevar N, Komel R. Proteomic approaches in 949.
biomarker discovery: new perspectives in cancer diag- [34] Huang Y-J, et al. Concordance of two approaches in
nostics. Sci World J. 2014;2014:260348. monitoring of minimal residual disease in B-precursor
[15] Lopez Villar E, et al. Proteomics-based discovery of bio- acute lymphoblastic leukemia: fusion transcripts and
markers for paediatric acute lymphoblastic leukaemia: leukemia-associated immunophenotypes. J Formos
challenges and opportunities. J Cell Mol Med. 2014;18 Med Assoc. 2017;116(10):774–781.
(7):1239–1246. [35] Schrappe M. Detection and management of minimal
[16] Malm J, et al. Developments in biobanking workflow residual disease in acute lymphoblastic leukemia.
standardization providing sample integrity and stab- ASH Educ Program Book. 2014;2014(1):244–249.
ility. J Proteomics. 2013;95:38–45. [36] Parikh S, Uparkar U. Assessment of minimal residual
[17] Yip C, Garcia A. Exploring the potential of platelet pro- disease in childhood acute lymphoblastic leukemia. J
teomics in children. Proteomics Clin Appl. 2014;8(11– Appl Hematol. 2016;7(2):47–53.
12):807–812. [37] Gaipa G, et al. Drug-induced immunophenotypic
[18] Gonzalez-Gonzalez M, et al. Genomics and proteomics modulation in childhood ALL: implications for
approaches for biomarker discovery in sporadic color- minimal residual disease detection. Leukemia.
ectal cancer with metastasis. Cancer Genomics 2004;19:49.
Proteomics. 2013;10(1):19–25. [38] Hasan AB, et al. Folia Med (Plovdiv). 2016;58(1):
[19] Adan A, et al. Flow cytometry: basic principles and 28–35.
applications. Crit Rev Biotechnol. 2017;37(2):163–176. [39] Theunissen P, et al. Standardized flow cytometry for
[20] Chiaretti S, Zini G, Bassan R. Diagnosis and subclassifi- highly sensitive MRD measurements in B-cell acute
cation of acute lymphoblastic leukemia. Mediterr J lymphoblastic leukemia. Blood. 2017;129(3):347–357.
Hematol Infect Dis. 2014;6(1):e2014073. [40] Karawajew L, et al. Minimal residual disease analysis by
[21] van Dongen JJ, et al. Euroflow antibody panels for stan- eight-color flow cytometry in relapsed childhood acute
dardized n-dimensional flow cytometric immunophe- lymphoblastic leukemia. Haematologica. 2015;100
notyping of normal, reactive and malignant (7):935–944.
leukocytes. Leukemia. 2012;26(9):1908–1975. [41] Abaza HM, et al. Neuropilin-1/CD304 expression by
[22] Denys B, et al. Improved flow cytometric detection of flow cytometry in pediatric precursor B-acute lympho-
minimal residual disease in childhood acute lympho- blastic leukemia: A minimal residual disease and
blastic leukemia. Leukemia. 2013;27(3):635–641. potential prognostic Marker. J Pediatr Hematol Oncol.
[23] Kalina T, et al. Euroflow standardization of flow cyt- 2018;40(3):200–207.
ometer instrument settings and immunophenotyping [42] Bhattacharjee M, et al. Synovial fluid proteome in rheu-
protocols. Leukemia. 2012;26(9):1986–2010. matoid arthritis. Clin Proteomics. 2016;13:12.
[24] Gaipa G, et al. Detection of minimal residual disease in [43] Malchow S, et al. Quantification of cardiovascular
pediatric acute lymphoblastic leukemia. Cytometry B disease biomarkers in human platelets by targeted
Clin Cytom. 2013;84(6):359–369. mass spectrometry. Proteomes. 2017;5(4).
[25] Coustan-Smith E, et al. Immunological detection of [44] Guerrera IC, Kleiner O. Application of mass spec-
minimal residual disease in children with acute lympho- trometry in proteomics. Biosci Rep. 2005;25(1–2):71–
blastic leukaemia. Lancet. 1998;351(9102):550–554. 93.
[26] Basso G, et al. New methodologic approaches for [45] Fenn JB, et al. Electrospray ionization for mass spec-
immunophenotyping acute leukemias. trometry of large biomolecules. Science. 1989;246
Haematologica. 2001;86(7):675–692. (4926):64–71.
HEMATOLOGY 649

[46] Karas M, Hillenkamp F. Laser desorption ionization of complex profiling. Expert Rev Proteomics. 2005;2
proteins with molecular masses exceeding 10,000 (6):879–889.
daltons. Anal Chem. 1988;60(20):2299–2301. [66] Huang Y, Zhu H. Protein array-based approaches for
[47] Cristea IM, Gaskell SJ, Whetton AD. Proteomics tech- biomarker discovery in cancer. Genomics Proteomics
niques and their application to hematology. Blood. Bioinformatics. 2017;15(2):73–81.
2004;103(10):3624. [67] Sutandy FX, et al. Overview of protein microarrays. Curr
[48] Yates JR. 3rd mass spectral analysis in proteomics. Protoc Protein Sci. 2013: 1–21. Chapter 27: p. Unit 27 1..
Annu Rev Biophys Biomol Struct. 2004;33:297–316. [68] Stoevesandt O, Taussig MJ. Affinity proteomics: the
[49] Lundstrom SL, et al. Spotlight proteomics: uncovering role of specific binding reagents in human proteome
the hidden blood proteome improves diagnostic analysis. Expert Rev Proteomics. 2012;9(4):401–414.
power of proteomics. Sci Rep. 2017;7:41929. [69] Hartsink-Segers SA, et al. Aurora kinases in childhood
[50] Churchman ML, et al. Efficacy of retinoids in IKZF1- acute leukemia: the promise of aurora B as therapeutic
mutated BCR-ABL1 acute lymphoblastic leukemia. target. Leukemia. 2013;27(3):560–568.
Cancer Cell. 2015;28(3):343–356. [70] Irwin ME, et al. Small molecule ErbB inhibitors decrease
[51] Saha S, et al. 2DGE and DIGE based proteomic study of proliferative signaling and promote apoptosis in
malignant B-cells in B-cell acute lymphoblastic leuke- Philadelphia chromosome–positive acute lymphoblas-
mia. EuPA Open Proteom. 2014;3:13–26. tic leukemia. PLoS One. 2013;8(8):e70608.
[52] Cavalcante Mde S, et al. A panel of glycoproteins as [71] Alatrash G. Targeting cathepsin G in myeloid leukemia.
candidate biomarkers for early diagnosis and treat- Oncoimmunology. 2013;2(4):e23442.
ment evaluation of B-cell acute lymphoblastic leuke- [72] Khan M, et al. Cathepsin G is expressed By B cell acute
mia. Biomark Res. 2016;4:1. lymphoblastic leukemia and is an effective immu-
[53] Mirkowska P, et al. Leukemia surfaceome analysis notherapeutic target. Blood. 2017;130(Suppl 1):1438.
reveals new disease-associated features. Blood. [73] Heltemes-Harris LM, et al. Ebf1 or Pax5 haploinsuffi-
2013;121(25):e149–e159. ciency synergizes with STAT5 activation to initiate
[54] Xu G, et al. Label-free quantitative proteomics reveals acute lymphoblastic leukemia. J Exp Med. 2011;208
differentially expressed proteins in high risk childhood (6):1135–1149.
acute lymphoblastic leukemia. J Proteomics. [74] Schinnerl D, et al. The role of the Janus-faced transcrip-
2017;150:1–8. tion factor PAX5-JAK2 in acute lymphoblastic leuke-
[55] Zhu YP, et al. Preparation of Whole bone marrow for mia. Blood. 2015;125(8):1282–1291.
mass cytometry analysis of Neutrophil-lineage cells. J [75] van der Veer A, et al. Interference with pre-B-cell recep-
Vis Exp. 2019;148:10.3791/59617. tor signaling offers a therapeutic option for TCF3-
[56] Wierz M, et al. High-dimensional mass cytometry rearranged childhood acute lymphoblastic leukemia.
analysis revealed microenvironment complexity in Blood Cancer J. 2014;4:e181.
chronic lymphocytic leukemia. Oncoimmunology. [76] Shojaee S, et al. Erk negative feedback control enables
2018;7(8):e1465167. pre-B cell transformation and represents a therapeutic
[57] Samusik N, et al. Automated mapping of phenotype target in acute lymphoblastic leukemia. Cancer Cell.
space with single-cell data. Nat Methods. 2016;13 2015;28(1):114–128.
(6):493–496. [77] Wang TX, et al. Reversal of multidrug resistance by 5,5’-
[58] Bendall SC, et al. Single-cell mass cytometry of differen- dimethoxylariciresinol-4-O-beta-D-glucoside in doxor-
tial immune and drug responses across a human ubicin-resistant human leukemia K562/DOX. Indian J
hematopoietic continuum. Science. 2011;332 Pharmacol. 2013;45(6):597–602.
(6030):687–696. [78] Stam RW, et al. Association of high-level MCL-1
[59] Behbehani GK. Applications of mass cytometry in clini- expression with in vitro and in vivo prednisone resist-
cal Medicine: The Promise and Perils of clinical CyTOF. ance in MLL-rearranged infant acute lymphoblastic
Clin Lab Med. 2017;37(4):945–964. leukemia. Blood. 2010;115(5):1018–1025.
[60] Behbehani GK, et al. Mass cytometric functional [79] Huang YJ, et al. Reverse-phase protein array analysis to
profiling of acute myeloid leukemia defines cell-cycle identify biomarker proteins in human pancreatic
and immunophenotypic properties that correlate cancer. Dig Dis Sci. 2014;59(5):968–975.
with known responses to therapy. Cancer Discov. [80] Petit C, et al. Hypoxia promotes chemoresistance in
2015;5(9):988–1003. acute lymphoblastic leukemia cell lines by modulat-
[61] Ferrell Jr. PB, et al. High-dimensional analysis of acute ing death signaling pathways. BMC Cancer.
myeloid leukemia reveals phenotypic changes in per- 2016;16:746.
sistent cells during induction therapy. PLoS One. [81] Yang Y, et al. Wnt pathway contributes to the protec-
2016;11(4):e0153207. tion by bone marrow stromal cells of acute lympho-
[62] Fisher DAC, et al. Mass cytometry analysis reveals blastic leukemia cells and is a potential therapeutic
hyperactive NF Kappa B signaling in myelofibrosis target. Cancer Lett. 2013;333(1). .
and secondary acute myeloid leukemia. Leukemia. [82] Ghazavi F, et al. RPPA-based protein profiling reveals
2017;31(9):1962–1974. enhanced PI3 K/AKT/mTOR signaling in ETV6/RUNX1-
[63] Levine JH, et al. Data-driven phenotypic dissection of positive acute lymphoblastic leukemia patients with
AML reveals progenitor-like cells that correlate with low CD200 expression. Blood. 2016;128(22):890.
prognosis. Cell. 2015;162(1):184–197. [83] Guzmán-Ortiz AL, et al. Proteomic changes in a child-
[64] Lamble AJ, et al. Integrated functional and mass spec- hood acute lymphoblastic leukemia cell line during
trometry-based flow cytometric phenotyping to the adaptation to vincristine. Boletín Médico del
describe the immune microenvironment in acute Hospital Infantil de México. 2017;74(3):181–192.
myeloid leukemia. J Immunol Methods. 2018;453:44–52. [84] Kawahara R, et al. Integrative analysis to select cancer
[65] Mattoon D, et al. Biomarker discovery using protein candidate biomarkers to targeted validation.
microarray technology platforms: antibody-antigen Oncotarget. 2015;6(41):43635–43652.
650 A. F. CITALAN-MADRID ET AL.

[85] Quezada H, et al. Omics-based biomarkers: current [102] Gostissa M, et al. Conditional inactivation of p53 in
status and potential use in the clinic. Boletín mature B cells promotes generation of nongerminal
Médico del Hospital Infantil de México. 2017;74 center-derived B-cell lymphomas. Proc Natl Acad Sci
(3):219–226. USA. 2013;110(8):2934–2939.
[86] Tembhare Prashant R, et al. Evaluation of new markers [103] Steeghs EMP, et al. JAK2 aberrations in childhood B-cell
for minimal residual disease monitoring in B-cell pre- precursor acute lymphoblastic leukemia. Oncotarget.
cursor acute lymphoblastic leukemia: CD73 and CD86 2017;8(52):89923–89938.
are the most relevant new markers to increase the [104] Jerchel IS, et al. RAS pathway mutations as a predictive
efficacy of MRD 2016; 00B: 000–000. Cytometry Part biomarker for treatment adaptation in pediatric B-cell
B: Clin Cytometry. 2016;94(1):100–111. precursor acute lymphoblastic leukemia. Leukemia.
[87] Sherif L, et al. Cluster of differentiation 97 as a bio- 2018;32(4):931–940.
marker for the detection of minimal residual disease [105] Antonyak MA, Cerione RA. Ras and the FAK paradox.
in common acute lymphoblastic leukemia. Egypt J Mol Cell. 2009;35(2):141–142.
Haematol. 2017;42(3):81–87. [106] Wang X, et al. LRG1 promotes angiogenesis by modulat-
[88] Feist P, Hummon AB. Proteomic challenges: sample ing endothelial TGF-β signalling. Nature. 2013;499:306.
preparation techniques for microgram-quantity [107] Chaiwatanasirikul KA, Sala A. The tumour-suppressive
protein analysis from biological samples. Int J Mol function of CLU is explained by its localisation and
Sci. 2015;16(2):3537–3563. interaction with HSP60. Cell Death Dis. 2011;2:e219.
[89] Arques S. Human serum albumin in cardiovascular dis- [108] Wang S-J, et al. Overexpression of cysteine and
eases. Eur J Intern Med. 2018;52:8–12. glycine-rich protein 2 in bone marrow as a novel bio-
[90] Bene MC, Faure GC. CD10 in acute leukemias. GEIL marker in adult B-cell acute lymphoblastic leukemia.
(Groupe d’Etude Immunologique des Leucemies). Blood. 2016;128(22):5271–5271.
Haematologica. 1997;82(2):205–210. [109] Ebrahimi-Rad M, et al. Adenosine deaminase 1 as a bio-
[91] Al-Mawali A, et al. Incidence, sensitivity, and specifi- marker for diagnosis and monitoring of patients with
city of leukemia-associated phenotypes in acute acute lymphoblastic leukemia. J Med Biochem.
myeloid leukemia using specific five-color multipara- 2018;37(2):128–133.
meter flow cytometry. Am J Clin Pathol. 2008;129 [110] Asai D, et al. IKZF1 deletion is associated with a poor
(6):934–945. outcome in pediatric B-cell precursor acute lymphoblas-
[92] Bachir F, et al. Characterization of acute lymphoblastic tic leukemia in Japan. Cancer Med. 2013;2(3):412–419.
leukemia subtypes in Moroccan children. Int J Pediatr. [111] Yamashita Y, et al. IKZF1 and CRLF2 gene alterations
2009;2009:674801. correlate with poor prognosis in Japanese BCR-ABL1-
[93] Wang K, Wei G, Liu D. CD19: a biomarker for B cell negative high-risk B-cell precursor acute lymphoblastic
development, lymphoma diagnosis and therapy. Exp leukemia. Pediatr Blood Cancer. 2013;60(10):1587–
Hematol Oncol. 2012;1(1):36. 1592.
[94] Poe JC, et al. CD22 regulates B lymphocyte function in [112] Milani M, et al. Plasma Hsp90 level as a marker of early
vivo through both ligand-dependent and ligand-inde- acute lymphoblastic leukemia engraftment and pro-
pendent mechanisms. Nat Immunol. 2004;5(10):1078– gression in mice. PLoS One. 2015;10(6):e0129298.
1087. [113] Zuehlke A, Johnson JL. Hsp90 and co-chaperones twist
[95] Luger D, et al. Expression of the B-cell receptor com- the functions of diverse client proteins. Biopolymers.
ponent CD79a on immature myeloid cells contributes 2010;93(3):211–217.
to their tumor promoting effects. PLoS One. 2013;8 [114] Xu Y, et al. Overexpression of MALT1-A20-NF-kappaB in
(10):e76115. adult B-cell acute lymphoblastic leukemia. Cancer Cell
[96] Martin-Martin L, et al. Immunophenotypical, morpho- Int. 2015;15:73.
logic, and functional characterization of maturation- [115] Obro NF, et al. Identification of residual leukemic cells
associated plasmacytoid dendritic cell subsets in by flow cytometry in childhood B-cell precursor acute
normal adult human bone marrow. Transfusion. lymphoblastic leukemia: verification of leukemic state
2009;49(8):1692–1708. by flow-sorting and molecular/cytogenetic methods.
[97] Vardiman JW, et al. The 2008 revision of the World Haematologica. 2012;97(1):137–141.
Health organization (WHO) classification of myeloid [116] Wang W, et al. The application of CD73 in minimal
neoplasms and acute leukemia: rationale and impor- residual disease monitoring using flow cytometry in
tant changes. Blood. 2009;114(5):937–951. B-cell acute lymphoblastic leukemia. Leuk
[98] Kong Y, et al. CD34 + CD38 + CD19+ as well as CD34 + Lymphoma. 2016;57(5):1174–1181.
CD38-CD19+ cells are leukemia-initiating cells with [117] Shi L, et al. Discovery and identification of potential
self-renewal capacity in human B-precursor ALL. biomarkers of pediatric acute lymphoblastic leukemia.
Leukemia. 2008;22(6):1207–1213. Proteome Sci. 2009;7:7.
[99] Djokic M, et al. Overexpression of CD123 correlates [118] Good Z, et al. Single-cell developmental classification
with the hyperdiploid genotype in acute lymphoblastic of B cell precursor acute lymphoblastic leukemia at
leukemia. Haematologica. 2009;94(7):1016–1019. diagnosis reveals predictors of relapse. Nat Med.
[100] Herzog S, Reth M, Jumaa H. Regulation of B-cell pro- 2018;24(4):474–483.
liferation and differentiation by pre-B-cell receptor sig- [119] Sarno J, et al. SRC/ABL inhibition disrupts CRLF2-driven
nalling. Nat Rev Immunol. 2009;9(3):195–205. signaling to induce cell death in B-cell acute lympho-
[101] Forero-Castro M, et al. Mutations in TP53 and JAK2 are blastic leukemia. Oncotarget. 2018;9(33):22872–22885.
independent prognostic biomarkers in B-cell precursor [120] Bendall SC, et al. Single-cell trajectory detection
acute lymphoblastic leukaemia. Br J Cancer. 2017;117 uncovers progression and regulatory coordination in
(2):256–265. human B cell development. Cell. 2014;157(3):714–725.

You might also like