Circulating Biomarkers for B-Cell Progenitor Acute Lymphoblastic Leukemia: Comparison
Please note this is a comparison between Version 1 by Margarita L Martinez-Fierro and Version 2 by Alfred Zheng.

Acute lymphoblastic leukemia (ALL) is a hematological disease characterized by the dysfunction of the hematopoietic system that leads to arrest at a specific stage of stem cells development, suppressing the average production of cellular hematologic components. BCP (B-cell progenitor)-ALL is a neoplasm of the B-cell lineage progenitor. BCP-ALL is caused and perpetuated by several mechanisms that provide the disease with its tumor potential and genetic and cytological characteristics. These pathological features are used for diagnosis and the prognostication of BCP-ALL. The BCP-ALL diagnostic protocol is well established. Firstly, it is necessary to demonstrate ≥ 20% lymphoblasts in bone marrow (BM) based on a BPM. Second, a hematopathological review is performed; it comprises a morphological assessment, and flow cytometric and genetic characterization.

  • biomarkers
  • circulating
  • acute lymphoblastic leukemia
  • BCP-ALL

1. Introduction

Leukemias are a group of hematological diseases characterized by an abnormal cell population suppressing the average production of cellular components of the hematopoietic system [1]. There are four main subtypes of leukemia that have been identified based on their evolution time and hematological lineage: acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), acute myelogenous leukemia (AML), and chronic myelogenous leukemia (CML) [2]. ALL and CLL are the most common leukemia types in children and adults, respectively [3][4][3,4].
ALL is a malignant disorder that occurs when typical lymphoid cell development malfunctions due to arrest at a specific stage of development; the name and classification and based on the dysfunctional stage [5]. ALL can develop from primitive precursor cells with multilineage potential from the two types of lymphoblasts: B or T. These cells proliferate uncontrollably in bone marrow (BM) and peripheral blood (PB) [1][6][7][8][1,6,7,8] and interfere with the functions of normal blood cells [8][9][8,9]. Of the two kinds of lymphoblasts, it is more common for ALL to originate from B-cell progenitor (BCP-ALL), representing about 80% of cases; the remaining 20% originate from T-cell progenitors (TCP-ALL) [10].
According to the World Health Organization (WHO), BCP-ALL is defined as a neoplasm of precursor lymphoid cells committed to the B cell lineage, typically composed of small-to-medium-sized blast cells with scant cytoplasm, moderately condensed to dispersed chromatin, and inconspicuous nucleoli [5]. BCP-ALL diagnosis involves clinical and laboratory tests. BCP-ALL symptoms indicate blast infiltration in the bone marrow, lymphoid system, and extramedullary sites and may include fatigue or lethargy, constitutional symptoms, dyspnea, dizziness, infections, and easy bruising or bleeding [11]. A BM aspirate is mandatory for laboratory tests to confirm the diagnosis, demonstrating ≥ 20% bone marrow lymphoblasts [12]. In addition, clinicians can examine cell morphology, immunophenotyping, and genetics and integrate these measures according to the classification established by the WHO [5]. BCP-ALL has multiple subtypes characterized by immunophenotype B-I (pro-B), B-II (Common), B-III (pre-B), and B-IV (B—mature Burkitt type) [5]. Recently, the nomenclature of leukemia with genetic abnormalities has shifted from focusing on cytogenetic alterations to molecular events, and new genetic entities have been added. The WHO classification in 2022 is based on clinical–biological entities defined by cytogenetic alterations that confer different prognoses. The United Kingdom ALL copy-number alteration (UKALL-CNA) classification has been incorporated, which distinguishes three genetic risk groups [13].
The peak incidence of ALL is from 2 to 5 years of age. It is rare in adults, but it can be found after 50 years of age. BCP-ALL constitutes approximately 80–85% of total cases of ALL [14][15][16][14,15,16]. Many factors affect the prognosis of BCP-ALL, such as age, the white blood cell (WBC) count, clinical features at diagnosis, cytogenetic abnormalities, pharmacodynamic and pharmacogenetic characteristics of normal patient cells, early response to therapy, and the results of the measurements of minimal/measurable residual disease (MRD: the presence of leukemic cells at specific time points in bone marrow or even in peripheral blood circulation) [17][18][19][20][21][22][17,18,19,20,21,22]. The prognostic implication of cytogenic abnormalities relates to poorly regulated signaling pathways [5][23][5,23], which can affect leukemogenesis and directly influences ALL development [24][25][24,25].
The above-mentioned factors help determine a “risk stratification” classification for BCP-ALL. This classification determines the treatment regimen and intensity that will be settled to treat BCP-ALL. This classification has contributed to a marked improvement in the prognosis of patients with leukemia [19][26][27][19,26,27].
As mentioned before, a BM puncture (BMP)/aspirate is needed to diagnose BCP-ALL. However, because of its invasive nature, potential side effects, and expensive procedures, there is a real need for alternative measures and molecular markers for the effective diagnosis and prognosis of pediatric patients with BCP-ALL [14][28][14,28]. Suitable substitutes for BMP are molecular biomarkers found in peripheral blood; collecting blood produces minimal trauma, and biomarkers can be measured relatively easy and early in the disease [29][30][31][29,30,31]. Nevertheless, the mentioned replacement must fulfill the biomarker standards.
In oncology, a biomarker is any measurable indicator that demonstrates the presence of malignancy, tumor behavior, prognosis, or responses to treatments [32]. Among the myriad of biological materials circulating in the bloodstream, the most promising biomarkers include circulating tumor cells, cell-free DNA (cf-DNA) and RNA, proteins and metabolites, and extracellular vesicles (EVs) [33].

2. Diagnostic Biomarkers for BCP-ALL

The BCP-ALL diagnostic protocol is well established. Firstly, it is necessary to demonstrate ≥ 20% lymphoblasts in BM based on a BPM. Second, a hematopathological review is performed; it comprises a morphological assessment, and flow cytometric and genetic characterization. Once these studies are completed, a diagnosis according to the WHO classification can be made [12]. The BCP-ALL classification integrates morphology, immunophenotyping, and genetics/cytogenetics. There are no morphological features to distinguish between the BCP-ALL and TCP-ALL. Nevertheless, some lymphoblast characteristics are relevant: scant cytoplasm, size, and shape, wide relatively dispersed chromatin, and nuclear, and nucleolar peculiarities [34][59]. The immunophenotyping in BCP-ALL show some markers as almost always positive, namely CD19, cCD79a, cCD22, CD22, CD24, PAX5, and TdT; CD20, CD34, CD13, and CD33 expression is variable. Finally, among the genetic abnormalities BCR—ABL1, KMT2A-rearranged, ETV6-RUNX1, hyperdiploidy, hypodiploidy, IGH/IL3, TCF3-PBX1, BCR-ABL1-like, and iAMP21 can be mentioned [5]. The following circulating biomarkers have shown promise for either the diagnosis or prognosis of the illness.

2.1. Proteins Type BCP-ALL Biomarkers

Proteins are helpful biomarkers because they have multiple crucial functions, making them central in biological systems [35][36][48,60]. In an individual manner, tumor necrosis factor α (TNF-α) is among the most studied biomarkers for ALL. TNF-α is a cytokine that induces apoptosis by inhibiting the activity of caspases [37][61]. Ahmed et al. evaluated its levels in serum from adult patients with ALL, comparing patients who received chemotherapy to those who did not (Table 1). Based on receiver operating characteristic (ROC) curve analysis, TNF-α showed good ALL diagnostic utility, an area under the ROC curve (AUC) of 0.94, a sensitivity of 91.7%, and a specificity of 100% [3]. These reports are consistent with the findings reported by Aref et al., who found an increase in TNF-α levels among patients newly diagnosed and in remission compared with controls [38][62]. Ahmed et al., was able to confirm this using an ROC curve, even though Aref’s cohort of patients had both BCP-ALL and BCP-CLL. These findings indicate that TNF-α can be a valuable tool in diagnosing ALL [3][38][39][3,62,63]. Table 1 summarizes the markers whose diagnostic values for ALL and/or BCP-ALL have been determined.
Table 1.
ROC values of diagnostic biomarkers for BCP-ALL.
Video Production Service