Open Access

Advancements in leukemia management: Bridging diagnosis, prognosis and nanotechnology (Review)

  • Authors:
    • Jingbo Li
    • Yingxue Wang
    • Chunli Dong
    • Lifu Luo
  • View Affiliations

  • Published online on: October 2, 2024     https://doi.org/10.3892/ijo.2024.5700
  • Article Number: 112
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Leukemia is a cancer that starts in blood stem cells in the bone marrow. Today, the proper diagnosis and prognosis of leukemia are essential in mitigating the morbidity and mortality associated with this malignancy. The advent of novel biomarkers, particularly those related to minimal residual disease, has paved the way for personalized therapeutic strategies and enables the quantitative assessment of patient responses to treatment regimens. Novel diagnostic and targeted drug delivery may be helpful for the improved management of leukemia. Genetic clinical parameters, such as chromosomal abnormalities, are crucial in diagnosing and guiding treatment decisions. These genetic markers also provide valuable prognostic information, helping to predict patient outcomes and tailor personalized treatment plans. In the present review, the studies on the diagnostic and prognostic parameters of leukemia were analyzed. The prognosis of leukemia was investigated in most of the studies, and the remaining were performed on diagnosis. The clinical and laboratory prognostic parameters were the most common, followed by diagnostic hematological parameters, diagnostic blood parameter studies, and diagnostic immunological parameters. Clinical and laboratory prognostic and hematologic parameters were the most extensively studied. The methods used to diagnose and prognose the leukemia cases in these studies were predominantly clinical hematology. Numerous surface proteins and receptors, including CD45, CD27, CD29, CD38, CD27, CD123, CD56 and CD25, react similarly in various kinds of leukemia, which are ideal for targeted drug delivery. Drug delivery to leukemia cells encounters several significant obstacles, including heterogeneity, that hinder the effectiveness of treatment. Nanocarriers play a critical role in targeted drug delivery for leukemia by enhancing the precision of treatments directed at surface proteins and receptors. Additionally, they can be functionalized with targeting drugs and antibodies to target specific tissues and cells.

Introduction

Leukemia refers to a category of blood malignancies characterized by the excessive production of abnormal blood cells and an underlying propensity for the bone marrow to serve as cancer's site of origin (1). Malignant cells use up healthy bone marrow cells to grow, leaving the body without enough platelets and normal white blood cells (WBCs) to prevent bleeding and fight infection (2-4). It is a somatic mutation-driven hematological clonal neoplasm (5-7). Neoplastic cells can self-replicate, develop, and feed progenitor cells such as hematopoietic cells, exhibiting similar behaviour (8-10). Leukemic unipotent stem cells can differentiate into phenotypes of adult blood cells (11,12). Certain forms of leukemia progress more slowly than others, while others begin in myeloid cells rather than lymphocytes. Leukemia types vary in prognoses and available diagnostics (13-15).

Leukemia is among the most prevalent cancers worldwide (16,17). Because of premature mortality, leukemia has a significant impact on developing nations. However, leukemia may be controlled in developed countries (18). The disease rapidly proliferates and can be classified as acute or chronic, myeloid or lymphoid (19). The most prevalent forms of leukemia are those that impact the myeloid and lymphoid systems, including acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL) (20).

What causes most leukemia subtypes remains unknown, even though various genetic and environmental risk factors have been identified. It is common practice to perform a bone marrow biopsy to get a final diagnosis, with the results informing various treatments, from chemotherapy to stem cell transplantation. Different types of leukemia have other outcomes (21). The fact that isolated leukemia stem cells differ from normal blood stem cells is a favorable indication for discovering a specific drug targeting them (22).

The present review highlighted methods and studies associated with leukemia diagnosis and prognosis. A systematic review was conducted to learn about the numerous types of leukemia and their associated gene signatures. The parameters used in diagnosis have the potential to enhance treatment efficacy and protect infected patients from developing chronic conditions. An assessment can inform the timing and course of treatment of prognostic factors at the time of diagnosis. Over time, researchers have examined several potential prognostic indicators. The present study discussed current leukemia diagnosis and prognosis factors, concentrating on the clinical and pathological. In the present review, the AML, ALL, CML and CLL categories to were used classify the numerous forms of leukemia.

Methods

Implementing comprehensive search strategies for leukemia research

Although the databases search engines automatically look for different forms of search terms, the terms for British or US English were still used. Whole or parts of the search operators (Boolean), truncation symbols and proximity operators were used to add additional layers of control over our search results. Search operators, also known as 'Boolean' operators (AND, OR, NOT), were used to combine terms in exact ways. Parenthesis was used to specify the order of the search operators. Truncation symbols (*, $, ?) were used to control the plural forms and variant spellings. Lastly, the proximity was used to specify the distance between the terms.

The search carried out was as the following: (((TS=('leukemia' OR 'hematologic malignancy' OR 'haematology malignancy' OR 'leukaemia' OR 'cancer of the blood' OR 'blood cancer')) AND TS=('prognosis' OR 'outcome prediction'))) AND TS=('*nano*'). There were 205 results from Science Citation Index Expanded (SCI-EXPANDED), Social Sciences Citation Index (SSCI), Arts and Humanities Citation Index (A&HCI), Conference Proceedings Citation Index - Science (CPCI-S), and Emerging Sources Citation Index (ESCI). One article was published in Chinese and another in Persian. Both articles were excluded from the present study. Only the articles published in English were evaluated. These articles were exported in several formats. Plain text, Endnote desktop, and Excel file formats were exported, and all the features of the Web of Science (https://mjl.clarivate.com/home) were included. Similarly, the PubMed database (https://pubmed.ncbi.nlm.nih.gov/) was searched using these credentials: ((=('leukemia'[Title/Abstract] OR 'hematologic malignancy'[Title/Abstract] OR 'haematology malignancy'[Title/Abstract] OR 'leukaemia'[Title/Abstract] OR 'cancer of the blood'[Title/Abstract] OR 'blood cancer'[Title/Abstract]) AND (=('prognosis'[Title/Abstract] OR 'outcome prediction'[Title/Abstract])) AND (nano*[Title/ Abstract]). The search produced a total of 120 articles. The search output was exported as a citation manager, and the file format nbib and Endnote 20.6 were used for further analysis. the PRISMA rules were followed for this systematic review and the PRISMA flow chart is shown in Fig. 1.

Systematic information extraction for consistent data analysis in leukemia studies

A standardized data extraction sheet was employed to ensure uniformity in capturing critical information from each study, including study characteristics, participant details, intervention and comparator specifics, outcome measures and methodology. The most current and comprehensive study was selected in cases where multiple studies provided similar findings. All identified studies underwent rigorous screening, with titles and abstracts first assessed for eligibility. Articles meeting the criteria were then subjected to full-text review, and exclusions were documented with specific reasons. The final data synthesis involved comparing and contrasting the extracted data to identify consistent themes and areas requiring further research.

Key findings from leukemia diagnostic and prognostic studies

Analyzing the outcome of the comprehensive search on leukemia

In the first round of analysis, 205 published articles were produced by the Web of Science Core Collection and 120 articles were produced from the PubMed database. A total of two articles were excluded from the WoS database and one from PubMed because they were published in languages other than English. In the end, 203 articles from Web of Sciences and 119 articles from PubMed fulfilled the initial screening requirements and were analyzed for improved understanding.

The second step involved classifying the selected papers according to the diagnostic, prognostic and distinctive clinical aspects, as well as related molecular genetics. The retrieved material covered the study's methodology, conclusions, minimal residual disease (MRD) status, and survival rates for pediatric, adolescent and young adult leukemia patients.

There were numerous criteria for diagnosing leukemia, and the present review shows which ones were more accurate and frequently used. As observed in Table I, these factors showed a variety of leukemia types, with CLL being the least prevalent. Numerous surface proteins and receptors, including CD33, CD45, CD27, CD29, CD38, CD27, CD123, CD56 and CD25, react similarly to parameters in various kinds of leukemia, as demonstrated in Fig. 2.

Table I

Detail of all parameters in leukemia.

Table I

Detail of all parameters in leukemia.

Sr. no.First author/yearLeukemia typeParametersAge (years)Value of risk(Refs.)
1Kassahun et al, 2020AMLHematological15-4951.60%(23)
2Jahic et al, 2017AMLPrognostic<5586.40%(24)
3Zhang et al, 2009ALLClinical prognostic25-5020-30%(25)
4Kardum-Skelin et al, 2008Chronic leukemicClinical and laboratory prognostic>60>70%(26)
5Othieno-Abinya et al, 2016CMLHematologicalNR46%(27)
6Spiers, 2001CGLHematological60-6555%(28)
7Sharma et al, 1992LeukemiaBloodNR51%(29)
8Germing et al, 2002CMMLPrognostic7015-24%(30)
9Tonorezos et al, 2013Childhood leukemiaMetabolic18-3731%(31)
10Haferlach et al, 2010CLLPrognostic genetic>6589.50%(32)
11Jaeger et al, 2012CMLBone metabolic4-1758%(33)
12Letestu et al, 2010CLLPrognostic29-8780%(34)
13Negrini et al, 2014CLLPrognostic>70NR(35)
14Rigolin et al, 2012CLLClinic biological63-6921.5-35.7%(36)
15Babusikova et al, 2009TLLPrognostic20-3044%(37)
16Joncourt et al, 1995LeukemiaDrug resistance15 and 7935%(38)
17Baumgartner et al, 2016AMLNutritional4657.10%(39)
18Sillaber et al, 2003CMLPathophysiology, diagnostic≥5090-95%(40)
19Zittoun et al, 1984AMLPrognostic value of cytologic30-652.50%(41)
20Piard et al, 2006ALL Pharmacokinetic2-1613.20%(42)
21Halonen et al, 2003ALLSerum Iron2.6-1714%(43)
22Prabhu et al, 1986CMLPrognostic12-6539%(44)
23Shinawi et al, 2012CLLBiological prognosticNR35%(45)
24Royer-Pokora et al, 1978ALVTransformation3023%-80%(46)
25Oliveira et al, 2001CLLClinical and laboratory45-86>30%(47)
26Hofmann-Lehmann et al, 1997FLVDisease progressionSeven weeks83%(48)
27Chiaretti et al, 2014CLLPrognostic608.6-15.9%(49)
28Li et al, 2019ALLIVIM11-7093%(50)
29Dhasmana et al, 2016LeukemiaHematological5-8045.10%(51)
30Omer et al, 2017AMLHematological3-10129.30%(52)
31Heintel et al, 2001CLLPrognostic30-6830%(53)
32Molica et al, 2007CLLBiological45-85NR(54)
33Levine et al, 1971CLL and CMLClinical27-6590%(55)
34Martell et al, 2002CLLHematologic37-8728%(56)
35Bergmann et al, 2007CLLPrognostic30-8019.50%(57)
36Sinisalo et al, 2004CLLHumoral immunity48-7920%(58)
37Browman et al, 1983ANLLPredictiveNR50%(59)
38Wieser et al, 2009AMLPrognostic8997-99%(60)
39Valet et al, 2003AMLImmunophenotypic, cytogenetic and clinical>53.849.8 and 32.8%(61)
40Valent et al, 2008CMLDiagnostic and prognosticNRNR(62)
41Zimm et al, 1986ALLBiochemical20-6550%(63)
42Yang et al, 2014Acute leukemiaMorphological>7596.80%(64)
43Olson et al, 2020LGLLHematological14-6530-40%(65)
44Srinivasan et al, 2020CLL Clinico-hematological43-8865%(66)
45Hallek et al, 2019CLLPrognostic>65Low risk 93.2%, very high 23.3%(67)
46Feller et al, 2004AMLMRD18-7697%(68)
47Mohammadi et al, 2020APLBlood10-8258.50%(69)
48Al-Bayaa et al, 2020AMLClinical11-5553% in females; 47% in males(70)
49Khazaal et al, 2019CMLDifferent blood18-8067.8% in males; 32.2% in females(71)
50Ogasawara et al, 2019CMLComplete blood count19-8995.20%(72)
51Michailov et al, 2019AMLMouse spermNR45%(73)
52Virk et al, 2019AMLVCS12-7756%(74)
53Vigón et al, 2020CMLImmunologicalNRCD56+ <4%; CD3-CD56+ CD16+ >7%;(75)
54Mahmood et al, 2018CLLClinic hematological>6567%(76)
55Kim et al, 2018Rat basophilicAllergicNR65%(77)
56Schillinger et al, 2018CMLStructural39-9190%(78)
57Khan, 2018AMLClinical and hematological3-6559%(79)
58 Podszywalow-Bartnicka et al, 2018LeukemiaLiveNR10-12%(80)
59Nägele et al, 2017ALLClinical laboratoryNR69%(81)
60Lekovic et al, 2017CMLPredictive18-7476.70%(82)
61Burhan, 2017LeukemiaBiochemical15-5575%(83)
62M.Elzubeir et al, 2016CLLPrognostic42-90100%(84)
63Wenn et al, 2015CMLPrognostic24-8310.50%(85)
64Trendowski et al, 2015LeukemiaCell cycle growth controlNR20%(86)
65Huang et al, 2015ALLPlatelet18-7957%(87)
66Rabizadeh et al, 2014Acute leukemiaAutomated blood countNR1.7-18.7%(88)
67Koltan et al, 2015ALLImmunological13-2215.9-99.8%(89)
68Aljamaan et al, 2015*pALLClinical3-780%(90)
69Rafiq et al, 2014CLLHematological, biochemical20-4595%(91)
70Pietras et al, 2014LymphoblasticImmune system15-2580%(92)
71Deschler et al, 2013AMLGeriatric60-8780%(93)
72Mauro et al, 2012CLL Clinicobiologic51-6170% vs. 49%(94)
73Dong et al, 2011CLLPrognostic>6080%(95)
74Onida et al, 2002CMLPrognostic20-8871%(96)
75Sagatys et al, 2012CLLPrognostic>50>30%(97)
76Jinlong et al, 2015AMLPrognostic15-5549%(98)
77Marcucci et al, 2011AMLPrognostic10-6090%(99)
78Labib et al, 2017ALLPrognostic<2->1080%(100)
79Neaga et al, 2021AMLPrognosticNR65%(101)

[i] NR, not reported; pALL, pediatric acute lymphoblastic leukemia; ALV, avian leukemia viruses; ANLL, acute non-lymphoblastic leukemia; FLV, feline leukemia virus; IVIM, intravoxel incoherent motion; LGLL, leukemia large granular lymphocyte; MRD, minimal residual disease; VCS, volume, conductance and scatter; APL, acute promyelocytic leukemia; TLL, T-cell acute lymphoblastic leukemia; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; ALL, acute lymphocytic leukemia; CML, chronic myeloid leukemia.

Critical assessment of diagnostic and prognostic developments in leukemia

In this section, the current advancements in leukemia diagnostics and prognostics were critically examined, and the significant strides and existing gaps within the literature were highlighted. The current analysis specifically investigated the recent advancements in molecular genetics and their crucial role in improving leukemia care. Focus was addressed on the latest diagnostic markers, treatment modalities and prognostic indicators that have the potential to transform personalized medicine in leukemia management. The key areas where recent technologies and therapeutic approaches have shown promise were identified, while also pointing out areas where further research is urgently needed to bridge the knowledge gaps. This critical assessment offers a comprehensive overview that can serve as a foundation for future studies, aiming to improve patient outcomes through more tailored and effective treatment strategies.

Baseline characteristics of included studies

Most studies investigated leukemia's prognosis, followed by leukemia diagnosis. The more common studies included clinical and laboratory prognostic parameters, diagnostic hematological parameters, diagnostic blood parameters studies and diagnostic immunological parameters. Clinical and laboratory prognostic and hematological parameters were the most extensively studied, while others have been studied in smaller numbers. The methods used to diagnose and prognose the leukemia cases in these studies were predominantly prediction analysis, such as clinical hematology and drug-related laboratory studies based on gene mutation (Fig. 3).

In the current review, leukemia's diagnostic and prognostic parameters were screened (Table I). The present systematic review provides the most comprehensive results about leukemia diagnosis and prognosis. There were several parameters for leukemia diagnosis and prognosis, but most studies have focused on hematological diagnosis.

As observed in Table I, AML has a progressive increase in the incidence rate from birth to ~50 years of age, a steady increase from ~50-54 years of age, more rapid growth from ~60-64 years of age, and a decline in the incidence rate in the oldest age groups (102,103). In women, the highest rates are observed among those ≥90, whereas in men, those aged 85-89 years-old have the highest rates (41).

Children under the age of five have the highest risk of developing ALL. After reaching its lowest point in the mid-20s, the risk gradually increases again, starting at age 50. In total ~4/10 individuals diagnosed with ALL are adults (50,104).

The rate of developing CML increases gradually from birth but more rapidly between the ages of 65-69. The highest mortality rates are among women aged 85-89 and men ≥90. In most age categories, females and males experience the same rate of occurrence (96,105,106).

CLL is recognized as the second most frequent kind of adult leukemia. The incidence rates of CLL increase with age, peaking between 45-49 years-old for men and gradually for women. The highest mortality rates are among women aged 85-89 and men ≥90. Generally, women have lower incidence rates than men throughout a wide range of ages (107).

Exploring diagnostic parameters in leukemia

There are numerous parameters for leukemia diagnosis and prognosis, but clinical diagnosis is more specific and commonly used. Diagnostic parameters are used to screen for leukemia, identify the cause or origin of the disease, and improve leukemia diagnosis to find an improved way to respond to therapy. As observed in Table I, imatinib and the detection of BCR-ABL1 and microRNA (miRNA), which are often performed using polymerase chain reaction (PCR), reverse-transcription quantitative PCR (RT-qPCR), and fluorescence in situ hybridization (FISH), are essential for the diagnosis of leukemia (107).

However, modern diagnostic methods such as FISH, flow cytometry (FCM) and RT-qPCR have been incorporated into the effort to define this entity within the context of clinical practice, as revealed in Fig. 4. Algorithms of varying complexity have been created to use these methods to determine leukemia (108,109). Because of its convenience, speed, low cost and overall reliability, FCM has been widely employed to detect overexpression in the diagnostic panel.

Assessing leukemia through blood diagnostics

The first line of defense in detecting leukemia is a blood test. Because normal blood tests are performed for various reasons aside from leukemia, a blood test that includes a complete blood count (CBC) and differentials is a crucial opportunity to screen for leukemia early (110). Some of the hematological parameters studied are hemoglobin <7.4±1.6 g/dl, red blood cells <2.84±0.32 millions/mm3, erythrocyte sedimentation rate (ESR) <96.5±26.8 mm/1st h, platelets <46.0±70.2x103/µl and WBCs <56.1±26.8x103/µl (29). The creatinine, total bilirubin, urea and ALT values are higher than the average local population, but the alkaline phosphatase level is markedly lower. Individuals with leukemia have high levels of WBCs, as observed in Fig. 5. The absolute basophil count (111) is a reliable method for detecting CML. An absolute basophil count field is an affordable and simple diagnostic test for the early identification of patients with CML (72).

The platelet-related parameters platelet count (PLT), mean platelet volume (MPV), platelet distribution width (PDW), platelet-large cell ratio (P-LCR) and plateletcrit (PCT) are measured using an automated hematology analyzer and whole-blood FCM (112).

Platelet parameter activation and dysfunction are observed in patients with ALL, and glycoprotein expression is used to assess the disease's impact. Future research should look at platelet function from various perspectives, including extending platelet parameters related to illness and investigating platelet parameters with ALL over time. A practical approach for the diagnosis and prediction of disease, and more accessible methods of platelet detection are developed in other studies (112). The mechanism of platelet dysfunction must be unraveled; strategies for the straightforward application of therapeutic, prophylactic platelet transfusions must be developed (87).

Evaluating leukemia via bone marrow diagnostics

A bone marrow biopsy is required to determine the most effective therapy for leukemia. Testing the bone marrow for leukemia cells is crucial since the disease begins in the bone marrow (113). Bone marrow that produces sufficient blood cells can be determined through a bone marrow aspiration or biopsy. These tests identify the cause of fevers and other mysterious illnesses and track the progression of blood and marrow cancers. It is possible to monitor a patient's response to treatment for leukemia by repeating the bone marrow tests utilized after diagnosis (67).

Most individuals with AML, ALL, CML and CLL have an imbalance in their blood, with too few adult red blood cells and platelets and too numerous immature WBCs. Myeloblasts (also known as blasts) are very early blood-forming cells generally not detected in the blood and may take up a large portion of the WBCs. The function of cells differs from fully developed WBCs. Although these results indicate leukemia, a specific diagnosis generally requires analyzing cells from the bone marrow (114).

Molecular-level diagnosis of leukemia

The presence of 20% blast cells in bone marrow or blood is one criterion for diagnosing leukemia; however, molecular genetics changes should be addressed, which may lower the blast requirement to <20% (115). Both the 2017 European Leukemia Net (ELN) and the 2019 National Comprehensive Cancer Network (NCCN) guidelines advise karyotyping and FISH testing for patients with AML (114,116). Sub-classification of AML, risk stratification and treatment decisions use different tests.

Conducting cytogenetic analysis for leukemia diagnosis

Human chromosomes and chromosomal disorders are the subjects of cytogenetics. Bone marrow samples screen for chromosomal abnormalities such as the Philadelphia (Ph) chromosome. Blood and bone marrow tests show diagnostic results, including a high WBC count and establishing a diagnosis of CML (61).

A human somatic cell contains 23 pairs of chromosomes. There is often a Ph chromosome in the bone marrow of those with CML, around 95% of the total. However, a small subset of the population does not carry the Ph chromosome while having CML-related symptoms. Individuals rarely fail to provide a positive result for the BCR-ABL1 fusion gene on chromosome 22. Similarly to those with a detectable Philadelphia chromosome, therapy for these patients is compelling.

Regular cytogenetic study is essential to the diagnostic process when dealing with a patient suspected of having acute leukemia. In total, ~50% of adults with AML are found to have chromosomal abnormalities. Recurrent genetic anomalies in AML are categorized by the World Health Organization (WHO) with a list of seven translocations, inversions and variants (114,116). The WHO classifies a patient as having 'AML with myelodysplasia-related features' if they have ≥20% blasts in their blood or marrow and multiple chromosomal abnormalities. Evaluation of at least 20 metaphase cells from bone marrow is advised for diagnosing a normal karyotype, and this number should be doubled to determine an aberrant karyotype. It is possible to diagnose a karyotype abnormality with a simple blood draw (117).

Utilizing karyotyping in leukemia diagnosis and prognosis

Karyotyping is an essential technique for diagnosing and understanding different types of leukemia, as it allows for the detection of chromosomal abnormalities that provide valuable insights into the prognosis and potential treatment options for patients (118,119). The process begins with collecting bone marrow or peripheral blood samples, with bone marrow aspirates commonly used due to their high concentration of dividing cells (120). These samples are then cultured in a laboratory setting to encourage cell division. This is crucial because karyotyping requires cells in the metaphase stage, where chromosomes are most visible under a microscope (117). The cells are fixed onto a microscope slide and stained, which highlights the chromosomal bands, allowing for the detailed analysis and identification of any genetic abnormalities present in the leukemia cells (121).

Karyotyping helps classify AML because it may detect the presence of multiple recurring translocations in AML leukemic cells. Complex chromosomal alterations can also be detected by karyotyping. Variations such as this are used to categorize patients with AML into risk groups. Karyotyping is currently a beneficial method. However, there are limitations to karyotyping. Before analyzing metaphase chromosomes, the cells in a sample must have proliferated. If the intended cells do not divide, researchers will not be able to examine their metaphase chromosomes (36). This means there is a chance of bias in the karyotyping outcome. A translocation can also involve a section of the chromosome that is too small to be detected by microscopic karyotyping. Cryptic chromosomal alterations are only detectable by an alternative technique such as FISH.

Karyotyping is pivotal in diagnosing and characterizing various types of leukemia. In ALL, karyotyping identifies fundamental translocations such as t(12;21)(p13;q22), which is linked with a favorable prognosis, and t(9;22)(q34;q11), known as the Philadelphia chromosome, which indicates a poorer prognosis (122). For AML, karyotyping detects abnormalities such as translocations t(8;21)(q22;q22) and inversions such as inv(16)(p13q22), which have significant implications for prognosis and treatment strategies (123). In CLL, common findings include deletions on chromosome 13q, which is associated with a relatively improved prognosis, and mutations on chromosome 17p, which are linked to a more aggressive disease course (124). Through these detailed chromosomal analyses, karyotyping not only aids in accurate diagnosis but also helps tailor individualized treatment plans and predict patient outcomes.

Applying FISH techniques in the diagnosis of leukemia

Translocation can be effectively detected using FISH. In FISH assays, specific DNA sequences known as probes are used to target particular regions of chromosomes. A DNA probe is used when its sequence is a perfect match for the DNA sequence in the target region of the genome. Probe DNA typically measures hundreds of kilobases. The conjugated fluorophore can be observed under a fluorescence microscope once bound to the specific region of genomic DNA of interest. As a means of chromosome identification, FISH probes can be designed to target only the centromeres. Locus-specific FISH probes allow for pinpointing a particular gene's location. This means that the FISH assay can detect both overt and cryptic chromosomal translocations. Metaphase and interphase targets are accessible for FISH. A higher degree of sensitivity than karyotyping is typical (94). It is possible to detect the BCR-ABL1 gene with FISH. Genomes are constructed from smaller pieces of DNA. Using FISH, the BCR and ABL1 genes can be located on chromosomes using colored probes that bind to the DNA.

Prenatal diagnostics have been revolutionized by multiplex ligation-dependent probe amplification (MLPA), a technique from the field of molecular genetics. MLPA uses up to 40 probes specific for a distinct DNA sequence (often exons of a particular gene of interest) to count the number of copies of each DNA sequence. The previous study has demonstrated that MLPA is a reliable approach for identifying genomic aberrations in CLL and has a significant relationship with FISH results in patients with CLL (125). The advantages of MLPA over FISH include speed and cost-effectiveness. While MLPA may simultaneously detect copy number alterations and point mutations in several target locations, FISH can only deliver information for a limited selection of genomic targets at a time.

Faster results from FISH assays are possible. The t(15;17) translocation may be identified with several different FISH techniques, including the dual-color dual-fusion FISH assay. FISH methods are superior to karyotyping for the early detection of acute promyelocytic leukemia (APL) due to their shorter cycle times. Both the 15q24 promyelocytic leukemia (PML) probe and the 17q21 retinoic acid receptor alpha (RARA) probe are used in a dual-color, dual-fusion test. The chromosomal breaks in both areas of study coincide. The assay's carefully thought-out structure allows for detecting PML-RARA. However, in circumstances where the translocation occurs between the RARA gene and other partner genes, the PML gene probe would not help detect the translocation using the dual-color dual-fusion FISH approach. The RARA dual-color break-apart FISH test was created for this reason (126). RARA dual-color break-apart FISH employs two probes for the RARA gene, each fluorescently tagged with a different fluorophore. For example, one RARA probe is labeled with a red fluorophore, whereas the other is marked with a green fluorophore. The binding sites of the two probes are the RARA gene center and the breakpoint (127). Such a generally applicable test design makes identifying RARA translocation with any number of partner genes possible. The FISH test has been widely used to diagnose APL. Fast results mean you can get a diagnosis and start treatment sooner, improving your chances of a positive outcome. However, the FISH assay has its limitations as well. While FISH can detect some cryptic translocations, it cannot identify the extremely rare cryptic RARA translocation. Due to this, RT-qPCR techniques have been developed to detect PML-RARA translocation. The RT-qPCR process can detect RARA translocations that are otherwise undetectable.

Cryptic chromosomal alterations can significantly impact the diagnosis and prognosis of leukemia. For instance, in CML, the Philadelphia chromosome, characterized by a translocation between chromosomes 9 and 22, may not always be detected by conventional karyotyping, especially if the rearrangement is complex or involves minor cryptic alterations. FISH techniques using probes specific to the BCR-ABL1 fusion gene can reliably identify these cryptic rearrangements, enhancing diagnostic accuracy and improving treatment planning (128). Similarly, in AML, small deletions or subtle translocations, such as those involving the FLT3 gene, may be missed by traditional karyotyping but are detectable through FISH analysis (128). Applying FISH and other advanced molecular techniques ensures that such cryptic alterations are not over-looked, providing a more comprehensive understanding of the genetic landscape of leukemia.

The clinical and prognostic significance of t(8;21) translocation in AML

T(8;21)-translocated AML produces a chimeric gene on the 8q-derived chromosome capable of transcription. Typically, karyotyping will be able to detect such a translocation. However, if cells containing the translocation do not multiply throughout the experiment or if the translocation is cryptic, a false-negative result may be obtained using karyotyping. To address this issue, the FISH assay has been adapted to be performed on cells in the interphase phase. Some hidden translocation can be discovered by interrogating interphase cells using the dual-color FISH test. This makes it a more sensitive method than karyotyping. A high total remission rate and a favorable prognosis are typical for the AML subtype identified by the t(8;21) translocation tests. Blasts in this kind of AML are not always >20%. However, the presence of t(8;21) should not rule out a diagnosis of AML, even if there are <20% blasts (129).

The role of FCM and immunohistochemistry in leukemia diagnostics

In FCM and immunohistochemistry, cell samples are treated with antibodies, which only bind to particular proteins on cells (130,131). Cells are viewed under a microscope in immunohistochemistry to determine whether or not the antibodies are attached to them (indicating the presence of the corresponding proteins). At the same time, FCM requires a specialized machine.

More straightforward techniques for platelet detection were created as a successful strategy for illness diagnosis and prediction (132,133). Quantitative measurements of PLT, MPV, PDW, platelet-large cell ratio (P-LCR) and platelet count (PCT) were taken using an automated hematology analyzer and whole blood FCM. Unraveling the cause of platelet failure and developing simple methods for administering therapeutic, prophylactic platelet transfusions are necessary (87). Future studies should examine platelet function from various angles, including extending the relationship between platelet parameters and disease and looking at platelet metrics over time.

Understanding immunophenotyping in acute leukemia

Acute leukemia's lineage involvement can be determined using immunophenotyping employing multiparameter FCM. No one appears to agree on what percentage of individuals with acute leukemia should be considered favorable for a marker. For most indicators, the expression threshold is set at ≥20% of leukemic cells, while field has applied for other features (17). Quantifying expression patterns of various surfaces and intracellular antigens is necessary for lineage assignment, diagnosing mixed phenotype acute leukemia (MPAL), and identifying aberrant immunophenotypes to determine MRD. A blast count determined by FCM should not replace morphologic assessment.

MPAL is a potential immunophenotypic presentation of acute leukemia that is positive for BCR-ABL1. These cases of leukemia should be treated as ALL with the addition of an ABL1 tyrosine kinase inhibitor (TKI) to the standard chemotherapy. CML blast crisis is considered as a possible secondary diagnosis in BCR-ABL1-positive leukemias (134). Identifiable immunophenotypic characteristics are linked to certain AMLs with recurring genetic abnormalities (41).

The T-cell antigen receptor (TCR) and the B-cell antigen receptor (BCR) are the main actors in the adaptive immune system. The TCR and the BCR identify pathogenic signals, activating the cells and allowing them to perform their protective role. The plasma membrane is the site of TCR and BCR signaling initiation, which later spreads into the cell to activate the cells and trigger an immune defense. However, improper T- and B-cell activation can harm the host and cause autoimmune diseases, immunodeficiency and malignancy (135).

Antibodies to T-cell antigens (CD2, CD3, CD4, CD5 and CD7) bind more cells than those to B-cell antigens (CD19, CD20, CD21, CD22, CD23 and CD24), which together make up 20-40% of the leukocytes in peripheral blood and have a 70:30 ratio. The CD4/CD8 ratio is lower in advanced CLL, and this simple immune function parameter appears to have a significant predictive value for patients, predicting the disease's clinical fate (136).

However, specific antibodies (CD9, CD11b, CD13, CD14, CD15, CD16, CD56, CD57, CD79a, CD95 and CD154) are sporadically negative for frozen cells, indicating either poor binding affinity or the loss of these surface antigens due to shedding or epitope degradation (137). Morphology, immunophenotype, cytochemistry and karyotype are combined to diagnose leukemias and lymphomas. The antigens CD19, CD20, CD21, CD22, CD23, CD24, CD25 and CD37 show the best differentiation between CLL and regular peripheral blood leukocytes (138). The results from the microarray for the expression of 48 CD antigens agree with those from FCM. The microarray allows for thorough immunophenotyping and soluble, fluorescently labeled antibodies can further define the whole cells caught on antibody dots (139).

A low CD4/CD8 ratio is characterized by advanced CLL. The clinical presentation of CLL may be predicted with a high degree of accuracy using this relatively uncomplicated indicator of immunological function.

PCR for leukemia detection and monitoring

This is a highly sensitive assay, capable of detecting even trace amounts of leukemia cells. The process is effective because it raises the DNA concentration in the sample, making it easier to observe. This analysis can detect subtle chromosomal alterations that other methods miss.

A PCR or a DNA test can detect the BCR-ABL fusion gene and other molecular abnormalities. In addition, PCR diagnostics could be utilized to check the efficacy of treatment. This test, for instance, is so sensitive that it can identify even a single abnormal cell among a million normal ones. Both blood and bone marrow cells can be used in this analysis. The PCR can confirm a diagnosis of CML. Testing for the presence of BCR-ABL gene copies after treatment is also helpful. When this gene is duplicated, leukemia remains even if no visible cancer cells are current (72).

A follow-up evaluation may be able to quantify disease-specific chromosomal or molecular abnormalities. However, most documented aberrations lack an examination of the link between the disease load in CR and the clinical prognosis. A further drawback of PCR for MRD detection is the lack of standardization and test variability (71).

In this study, genetic abnormalities are analyzed by using multiplex ligation-dependent probe amplification. In a single experiment, the polymerase chain reaction-based method known as MLPA can concurrently identify point mutations, copy number alterations, and perform DNA methylation in up to 50 genomic DNA sequences (140).

RT-qPCR

RT-qPCR is the most precise and sensitive approach for determining how much BCR-ABL1 gene is present in blood and bone marrow samples. Even if the Ph chromosome is absent in blood or bone marrow cells, a single CML cell can be recognized among 100,000-1,000,000 normal cells using cytogenetic testing. Assuming the patient's CML responds to treatment, qPCR testing is advised every three months or two years.

Routine molecular diagnostics testing requires a bone marrow (and blood) sample. The ideal situation would involve the extraction of DNA and RNA and the storage of live cells. However, if cell numbers were restricted, RNA extraction would be prioritized since it lends itself to molecular screening for leukemia-associated mutations. Del 17p was highly prevalent among patients with CLL who visited the Armed Forces Institute of Pathology (Rawalpindi, Pakistan). Not all of these individuals, however, required treatment. However, introducing promising new medications may enhance their overall survival periods (76).

The RT-PCR can assist in diagnosing common gene fusions, including RUNX1-RUNX1T1, CBFB-MYH11, MLLT3-MLL and DEK-NUP214. RT-PCR can identify these rearrangements if chromosomal morphology is weak or marrow morphology is typical. Still, there are no cytogenetic abnormalities (109). The NPM1, FLT3, CEBPA and MLL (myeloid/lymphoid or mixed-lineage leukemia, Drosophila) (141), NRAS (neuroblastoma RAS viral (v) oncogene homolog) (142), and WT1, KIT (v) Hardy-Zuckerman 4 oncogenes (143) of feline sarcoma mutations in these genes vary considerably amongst cytogenetic groupings. Alleles of AML produced by NPM1 or CEBPA mutations are considered active entities. Patients with cytogenetically normal AML (CN-AML) who receive other than low-dose chemotherapy or optimal supportive care should have these three mutations examined, even though testing is not necessary outside clinical trials (144).

A molecular diagnosis using RT-PCR can be helpful for common gene fusions like RUNX1-RUNX1T1, CBFB-MYH11, MLLT3-MLL and DEK-NUP214. If the quality of the chromosomal morphology is low or the marrow morphology is typical, RT-PCR can detect these rearrangements (145). The rate at which these mutations occur varies greatly amongst cytogenetic groupings - the WHO now recognizes AML alleles produced by mutations in NPM1 or CEBPA as distinct entities. In addition to FLT3 mutations, screening patients for these two signs in treatment trials might be helpful. Although testing for these three mutations is not presently deemed necessary outside clinical trials, the panel recommends doing so for patients with cytogenetically normal AML (CN-AML) who receive treatment other than low-dose chemotherapy or appropriate supportive care.

Identifying key prognostic parameters in leukemia

Prognosis is all patient characteristics used to estimate recovery or a disease's chance of recurrence. A prognostic parameter is the patient's malignancy or a trait to be considered while determining the prognosis. A prognostic or predictive factor influences the outcome of a particular cancer treatment. The decision on a course of treatment and a prediction is often made in conjunction with predictive and prognostic criteria. Leukemia is prognosed using factors such as age, WBC count and genetic abnormalities, among others. Age, WBC count, Immunoglobulin Variable Heavy Chain gene (IgVH) status, TP53 deletion and miRNA studies are the established factors with the most established prognostic importance. Almost every subtype of leukemia has prognostic cytogenetic variables, particularly acquired mutations that, if identified, allow for the definition of the optimal therapy for a particular patient.

Although several pre-treatment features have been shown to have predictive value in leukemia, only a few risk factors have surfaced in ongoing leukemia studies with intensive chemotherapy regimens. These include the patient's age, starting WBC count, karyotype and immunophenotype. Variables used to assess the likelihood of a patient surviving a malignant tumor are associated with clinical outcomes in either a positive or negative way. Predictive factors determine the effectiveness of a treatment in a particular medical setting. They may favor one therapy over another prognostic factor. The prognostic impact can be fine-tuned, and therapy selections can be made with the help of scoring systems that combine numerous prognostic indicators with an independent relationship with outcome criteria.

The impact of age on leukemia prognosis and treatment outcomes

The patient's diagnosis age is the most important prognostic factor in leukemia. Patients with AML <30 years-old performed worse than those aged ≥50. In seniors with AML, comorbidities and probable side effects against rigorous cytotoxic treatment are more prevalent, as are impaired performance status, secondary AML following antecedent myelodysplastic syndrome or cytotoxic therapy, and unfavorable chromosomal abnormalities (98). The patients' age remains a significant independent prognostic factor, even once these other characteristics are considered (146). Even with aggressive chemotherapy, the outlook is poor for patients aged ≥65, regardless of cytogenetic risk. Various aspects of the disease's biology have been implicated in the differential prognoses between adults and children. The ability to tolerate a specific treatment varies from one individual to the next. The chemotherapy drugs are metabolized differently depending on age. Methotrexate polyglutamate accumulates at far lower levels in adult B-lineage blasts with ALL than in children with the same condition (100). Most individuals with CML are adults. A total of 1/1000.000 children >10 years of age will contract leukemia. In total, ~1/100.000 adults will have this condition. All types of AML, CML and CLL, as well as their parameters and risk values are summarized in Table I.

Research shows that leukemia can develop at any age. In general, adults ≥60 years-old have a higher risk of developing leukemia. Both AML and CLL are more common in adults. Every year, ~3,811 children are diagnosed with leukemia. When it comes to childhood cancers, ALL is by far the most common type. Any element that raises an individual's probability of acquiring leukemia is considered a 'risk factor'. In most cases of leukemia, the causes or risk factors are unknown (147). For specific patients' most accurate risk categorization, research is needed to determine how to combine variables showing more significant angiogenesis with other prognostic markers, such as cytogenetic anomalies (148).

The importance of WBC count and bone marrow histology in leukemia prognosis

The WBC count tests are pivotal in diagnosing and characterizing different types of leukemia, with distinct patterns observed across the various forms (149-151). In ALL, there is typically an elevated WBC count due to an increase in lymphoblasts, although some cases may present with normal or decreased counts. At the same time, bone marrow histology typically shows a high number of lymphoblasts, immature lymphoid cells that dominate the marrow and replace normal hematopoietic elements (152), while lymphoblasts are generally small to medium-sized, with scant cytoplasm and prominent nuclei. Similarly, AML often shows a high WBC count with a significant number of myeloblasts, yet normal or low counts are also possible in some patients (114). The marrow is infiltrated by myeloblasts and immature myeloid cells featuring a high nucleus-to-cytoplasm ratio, and may include Auer rods and needle-like cytoplasmic inclusions. By contrast, CLL is generally characterized by a very high WBC count due to the accumulation of small, mature-appearing lymphocytes in the bone marrow (124). With their condensed chromatin and uniform appearance, these cells crowd out normal hematopoietic cells, often resulting in hypercellularity.

For CML, an elevated WBC count is observed, along with an increase in granulocytes such as neutrophils, eosinophils and basophils; this condition is also marked by the presence of the Philadelphia chromosome or the BCR-ABL fusion gene (153). In CML, bone marrow histology shows marked hypercellularity with an increased number of granulocytic precursors at all stages of maturation, as well as dysplastic megakaryocytes. However, the overall architecture of the bone marrow is preserved. Hairy Cell Leukemia (HCL) can result in a low WBC count, as the abnormal cells accumulate in the bone marrow and disrupt normal blood cell production (154). The bone marrow typically appears fibrotic or presents a 'dry tap' in HCL due to the infiltration of hairy cells. These cells are recognized by their irregular cytoplasmic projections and are often positive for tartrate-resistant acid phosphatase enzyme, leading to pancytopenia as a result of marrow failure.

The white blood cell count at diagnosis is the strongest predictor of treatment effectiveness in leukemia. In total, 50-60% of all patients have a leukocyte count >10x109/l at diagnosis, and 10% have a leukocyte count >100x109/l. Leukocyte levels <10x109/l are also present in another 30-40%. Neutrophil absolute numbers are commonly relatively low. Complete remission and the length of time spent in remission strongly correlate with leukocyte counts. The importance of this pretreatment step cannot be understated in AML and ALL since long-term remissions are unusual in patients presenting with high WBC counts using standard chemotherapy regimens (56).

However, some evidence suggests that, with the help of modern chemotherapy, a high WBC count at diagnosis in ALL is not always indicative of a poor prognosis. Also associated with a poor prognosis is having more than 80% of blasts in circulation. There is a correlation between leukocyte counts and the fraction of circulating blasts, although this has been demonstrated to have an independent prognostic value (155).

Evaluating clinical and laboratory prognostic parameters across leukemia types

All blood samples were routinely examined under a microscope for years, allowing the medical community to amass substantial knowledge about how cells change in various disease states. Laboratory workloads and economic demands have risen over the past few decades. The use of morphologic data for diagnosis has been rapidly declining along with the development of automated cell counters that can report a CBC. Only a small percentage of blood samples are examined under a microscope today (156).

A poor prognosis was observed in patients with either an elevated lactate dehydrogenase (LDH) serum level or a high peripheral blood leucocyte count at diagnosis. The hemoglobin value, PCT, and fibrinogen plasma level at diagnosis affected different outcome indicators. Serum β2-microglobulin elevation was a poor prognostic indicator in patients with AML >65 years-old. However, compared with age and cytogenetic risk, these characteristics have a relatively minor impact on prognosis (157).

Plasma, fibrinogen, bleeding time and LDH activity increased, while hemoglobin, hematocrit and PCT decreased (158). Despite having lower hemoglobin, hematocrit and PCT, patients with AML had increased plasma viscosity, ESR, fibrinogen, leukocyte count, bleeding time and LDH activity. Increased bleeding times were observed in patients with CML with high levels of fibrinogen, plasma viscosity and ESR. The filtering ability of the red cells was consistent across all conditions. The absolute basophil count is an effective screening tool for chronic myeloid leukemia. The total basophil count is a simple, low-cost and rapid diagnostic tool for the early detection of patients with CML (72).

Careful clinical assessment, together with clinical stages according to Rai and Binet, CLLPD subtype and progression variables, is a crucial link for creating a tailored therapy approach and an essential step in determining the degree and course of the disease. In contrast to 'researcher-dependent' parameters, laboratory parameters (hematologic and biochemical) obtained by straightforward venipuncture are objective quantitative measures that improve the use of these indicators as risk factors in CLL (26).

The role of miRNA patterns in determining clinical prognostic parameters for leukemia

The miRNAs have been shown to provide therapeutic guidance for patients with pediatric leukemia who need risk-adapted treatment (25). Oncogenic miRNAs are associated with a poor prognosis (159), whereas tumor suppressor miRNAs, which prevent proto-oncogene expression, are associated with a favorable prognosis (160). The miRNA expression could be a more effective marker for determining tumor origin and prognosis than traditional protein-coding gene arrays. A miRNA profiling study focusing on pancreatic tissues revealed that the expression of just 25 miRNAs can differentiate between pancreatic adenocarcinoma, chronic pancreatitis and normal pancreatic tissue (160,161). MiRNAs are being considered as treatment options for several malignancies, including AML, and biomarkers for diagnosis and prognosis. Experimental, pre-clinical techniques and synergistic activity with chemotherapeutic drugs demonstrated viable therapeutic targets, as revealed in Fig. 6. The development and eventual clinical application of new medicines with mimic or suppressive effects gave way to the therapeutic targeting of miRNAs in the future (101).

Recent research has demonstrated the role of aberrantly expressed miRNAs in the development and spread of cancer. The classification, diagnosis, prognosis, treatments and biomarkers of human cancers can all be based on their expression profiles. Microarray-based technology for miRNA expression profiling has revealed new information about the biology of some adult CLL and AML cases (162). Calin et al (163) found that CD5+ and CLL cells express miR-15a and miR-16-1 differentially. MiR-15a and miR-16-1 were linked to disease progression and prognostic factors and triggered apoptosis by targeting BCL2 (163). Mi et al (164) found 27 differentially expressed miRNAs between ALL and AML. MiRNAs can diagnose acute leukemia in children (164). Microarrays are a successful way to research miRNA profiling, which helps uncover oncogenes or tumor suppressors involved in disease development.

Notably, several miRNAs change in expression across pediatric and adult instances, indicating the existence of a miRNA pattern unique to children. In adult patients with AML, miR-146 is downregulated, whereas miR-146a is upregulated (162). Contrary to a prior study that found miR-335 downregulated in 27 adult patients with AML, it was upregulated in pediatric M1 patients (165). According to Li et al (166), adult patients with APL had elevated miR-100.

Strong evidence came from the high-throughput sequencing data's comparatively high accuracy. Because of the relatively limited sample size, a more thorough examination should be conducted before applying them to the risk stratification of patients with CN-AML. These findings provided insightful investigation into the role and clinical utility of miR-188-5p and let-7a-2-3p. Their prognostic significance and related aberrantly expressed genes and miRNAs can further our knowledge of the internal workings of leukemogenesis and develop new CN-AML therapies (98).

Most patients with ALL are now treated with systemic therapy and protocol-based treatments. However, 20-30% of patients still pass away following treatment. Early relapses are primarily to blame for dismal survival rates of patients with ALL. Such patients continue to receive inadequate care. Therefore, research into the molecular pathophysiology of leukemia in relapsed patients may offer new information about their prognosis (25). It was concluded from the aforementioned study that children who experienced ALL relapse displayed a unique miRNA expression pattern, which included high levels of miRNA expression. Testicular relapse has all but disappeared thanks to improved ALL treatment. However, pediatric ALL therapy is still hampered by controlling central nervous system (CNS) recurrence (167). Therefore, it was concluded that miRNA profiles are responsible for the CNS relapse of juvenile ALL. The miRNA cascade may be employed as a biomarker to identify early pediatric ALL regressions in the CNS. According to miRNA target prediction, some irregularly generated miRNAs may target genes essential in neuron function and neurotransmitter synthesis.

MiRNAs inhibit translation and promote mRNA breakdown to regulate gene expression post-transcriptionally. They modulate biological processes and regulate hematopoietic differentiation. MiRNA expression correlates with AML prognosis (101). Post-transcriptional gene expression regulators regulate development, cell differentiation and cell signaling. MiRNAs modulate gene expression patterns in numerous illnesses, decreasing carcinogenic genes in leukemia. In Figs. 7 and S1 it is depicted how various enzyme complexes act at the transcriptional and post-transcriptional stages to produce mature miRNAs.

Exploring the role of cytogenetics in leukemia prognosis

The clinical outcomes for individuals with AML following intensive treatment are most strongly associated with cytogenetic risk, as was first shown in 1982. A favorable risk aberration is a balanced translocation involving the core binding factor (CBF) transcription factor complexes, such as t(8;21), inversion (16), or t(16;16) (98). The definition of high-risk abnormalities is more contentious. A total of >5,800 patients with AML were included in the most extensive study to examine cytogenetic abnormalities and their association with prognosis. High-risk cytogenetic aberrations discovered in that study include abn(3q) (excluding t(3;5)(q25;q34)), inv(3)(q21q26)/t(3;3) (q21;q26), abn(5q)/del(5q), -5, -7, abn(7q)/del(7q), t(11q23) (excluding t(9;11)(p21-22;q23), and t(11;19)(q23;p13)), t(9;22) (q34;q11), -17, abn(17p). Patients with at least three abnormalities (not counting benign variants) are considered to have a complex-aberrant karyotype, which is the most commonly accepted definition (66). Excluding cytogenetic characteristics from the model reduced negative outcomes and blurred the line between favorable and intermediate survival (168).

While peripheral blood samples are suitable for cytogenetic analysis, this technique is not always effective because CLL clones do not reliably divide in culture. The prevalence of common abnormalities has been reduced thanks to FISH research. FISH panels evaluate the most common cytogenetic aberrations in CLL, and these include del(17p13) [p53], del(11q23) [ATM], trisomy 12, and del(5q) (13q14). To rule out large-cell lymphoma, another CD5-positive B-cell malignancy, some panels evaluate for t(11;14) (49).

TP53, a tumor suppressor gene lost or mutated in numerous cancers, is deleted due to the 17p13 deletion. The standard protein p53, which is the product of TP53, binds to the p21 gene and increases p21 production. The p21 subsequently binds to CDK, the protein responsible for signaling cell division, inhibiting CDC's ability to do so and thus stopping the cell cycle from progressing. When p53 is absent, cells divide without control. Patients with CLL who have had their TP53 deleted tend to have a poor prognosis and a shorter overall survival than those who have not. Among 50 B-CLL clones without mutated IgVH, there are genetic abnormalities such as del (17p13) (134).

Utilizing Rai and Binet staging systems for leukemia prognosis

Rai et al (169) and Binet et al (170) established the gold standard CLL staging systems. These methods cannot detect stable or progressing sickness (171). The Rai and Binet staging systems affect survival of patients with CLL. The Rai CLL classification system is commonly used in North America. Lymphocytosis >15x109/l for four weeks, anemia with hemoglobin level <110 g/l, and thrombocytopenia of <100x109/l platelets define the five stages (from 0 to 4). This is on top of whether or not there is splenomegaly or lymphadenopathy. Stage 0 is the lowest possible danger in the Rai system, whereas stages 1 and 2 represent a moderate risk, and stages 3 and 4 mean extreme risk. CLL is classified into stages A, B and C according to the Binet staging approach, which is more often used in Europe. In addition to CLL in several sites (for example, head and neck, axilla, groin, palpable spleen, or clinically enlarged liver), the number of regions implicated and peripheral blood hemoglobin level and PCT are essential.

Reliable prognostic markers for individuals with chronic CLL have become increasingly important as treatment options have expanded in recent years. A prognostic score was constructed using multivariate analysis of 339 patients who were already in stage A at the time of diagnosis and had been thoroughly analyzed for traditional and contemporary predictive markers (34). Finally, a simple, rapid and cost-effective technique was suggested for reliable prognostication in stage A patients, who account for >80% of the CLL population. This enables clinicians to tailor follow-up individually.

Assessing leukemia prognosis through IgVH mutation analysis

The IgVH mutations are analyzed by comparing the DNA sequences of CLL B-cells to germline sequences, a process accomplished using PCR. DNA is considered to be mutated if its sequence deviates from the germline sequence by >2%. Patients with CLL with mutant IgVH have an improved prognosis because they are more likely to go without treatment for longer. Patients with the VH3-21 gene segment are an anomaly; they are members of the mutant group but act like unmutated clones with increased p53 dysfunction and shorter lifespans. CD38 expression, ZAP70 positivity, and del(17p) and del(11q) are associated with unmutated IgVH (11q23). The constraints of PCR testing are also present in IgVH mutation analysis, including the need for freshly collected samples (58). CD38 expression is associated with complex aberrant karyotypes, such as 13q deletions with mutated IgVH and TP53 deletions with unmutated IgVH. The parameters identify overlapping leukemia subgroups (172).

Understanding the prognostic implications of FLT3 mutations in leukemia

In total, ~30% of cases of AML have been linked to FLT3 mutations. While the FLT3 mutation is considered a driver mutation, it is not used to classify a specific form of AML. A poor prognosis is typically associated with FLT3 mutations. AML can now be treated with FLT3 inhibitors. Common FLT3 mutations include tyrosine kinase domain point mutations and FLT3 internal tandem duplicate (FLT3-ITD) repetitions (FLT3-TKD). The biological effects of these two sorts of mutations may be different. More evidence has emerged connecting FLT3-ITD with a poor prognosis. Guidelines for diagnosing and treating AML now consider the proportion of patients with the FLT3-ITD mutation compared with those with the FLT3 wild type (173). FLT3 mutations are discovered using several methods. One method amplifies FLT3 and then analyzes the PCR products using capillary gel fragments. The FLT3-ITD PCR products are longer than the wild-type. The peak's height inversely reflects the amount of mutant or wild-type PCR product. Peak height is used to determine the ratio of FLT3-ITD mutants to the wild type. This approach may be tweaked to find FLT3-TKD (174). The most common FLT3-TKD mutation is D835. Wild-type D835 has an EcoRV cutting site. EcoRV may quickly degrade FLT3 in its natural condition.

Meanwhile, a change to FLT3 D835 can render the protein inaccessible to EcoRV digestion. Using fragment analysis, the difference after the EcoRV digestion can be distinguished (175). To detect both FLT3-ITD and FLT3-TKD, a multiplex PCR and fragment analysis method was developed.

Evaluating MRD as a prognostic marker in leukemia

The timing of therapy initiation affects CLL progression since it depends on discovering prognostic variables at the diagnosis and the toxicity of different treatment regimens. However, permanent remission should be the ultimate objective. Determining MRD is becoming increasingly critical. High-precision, cutting-edge methods for detecting MRD include FCM and qPCR (176).

Patients whose therapeutic approaches have monoclonal antibodies may benefit most from the most recent test version. Patients with limited residual illness have a more reserved prognosis and a higher chance of relapse, but the use of additional targeted therapy remians debatable. MRD, defined as one CLL cell per 10,000 leukocytes, is a prognostic marker (177). The capacity to detect AML-MRD has been recognized as crucial for risk stratification and prognosis. MRD can be evaluated with either FCM or molecular testing. In this context, molecular diagnostics take center stage (178).

Mutations in a wide variety of genes have been linked to AML. Not all of these mutations can be used in MRD tests, however. NPM1 mutations and the fusion genes RUNX1-RUNX1T1, CBFB-MYH11 and PML-RARA can be MRD indicators. However, DNMT3A, ASXL1, and TET2 mutations are age-related and are not predictive of outcomes. That being said, it is possible that these mutations are not reliable molecular markers for AML MRD tests (178). Due to the clonal evolution of AML, mutations in genes are constantly being lost and gained during relapses (179).

The results of several molecular assays can be obtained depending on which molecular markers are of interest. A conventional qPCR is a valuable tool when a mutation in a single gene is chosen as an MRD marker. The NPM1 mutation can be checked with qPCR as an example. The quantitative analysis of NPM1 can now be performed using the cutting-edge field of digital PCR assay (180).

Balatzenko et al (181) evaluated the effects of BCR-ABL fusion types and the MDR1 gene on the hematological parameters of patients with CML. P-glycoprotein, expressed by the human MDR1 gene, exports molecules from cells and membranes to the extracellular environment. It protects cells from toxic metabolites. Only individuals with MDR1 overexpression showed a link between b3a2 and PCT (182). The scientists ascribed this rise to a signaling cascade between 3a2 and MDR1.

Role of imatinib in the prognosis of leukemia treatment

Numerous efforts have been made over the past 20 years to pinpoint molecular alterations that could cause cancer cells to resist cytotoxic medications (183). Multiple resistance mechanisms have been discovered in model systems or cell lines. Examples are the reduction in drug uptake or activation, enhancement of drug efflux or inactivation, alteration in target enzyme levels, enhanced repair of DNA damage, altered nucleotide pools, and avoidance of some drugs' inhibitory effects via salvage pathways. It is known that several pathways contribute to drug resistance in vitro, but their role in vivo is less understood (Fig. 8). There appears to be a variety of factors that contribute to drug resistance in vivo. To demonstrate this point, it is considered that drug resistance can develop despite using numerous drugs with different mechanisms of action. More in-depth studies are required on the molecular processes and phenotypic drug resistance patterns in tumor cells to improve tailor chemotherapy to individual patients. Imatinib is an effective medication for the first treatment of CML (82). It was designed to target BCR-ABL specifically. In addition to ABL, however, numerous off-target kinases are also impacted by the medication at plasma concentrations (184). The medication binds to the BCR-ABL protein tyrosine kinase ATP pocket, preventing ATP from binding and blocking the phosphorylation of substrates, as shown in Fig. 9 (185). The sustained response rates to drugs are comparable to the outstanding results reported for adults (186).

In all studies, children and adolescents were administered imatinib (IM) with few complications, including cerebral edema (187), miscarriages and birth abnormalities (188). IM treatment is linked to toxicity, with most side effects appearing within the first two years and potentially improving with continued treatment. Generally, toxicity is mild to moderate and manageable. Symptoms can include lethargy, rashes, weight gain due to fluid retention (notably infraorbital edema), and occassionally bone pain. Abnormal liver chemistry is possible and can rarely lead to liver failure. Some patients in CP starting at 400 mg/day may experience cytopenias within the first year, often leading to neutropenia, thrombocytopenia and occasionally anemia (188). Other cardiac adverse effects, including tachycardia, pericarditis, hypotension, and hypertension, collectively have an incidence of <1.0%. However, it is unknown how long CML responses will last and whether this medication may have long-term adverse effects in pediatric patients, particularly in different age groups (189).

Conclusion

Early diagnosis is critical for effective leukemia treatment, though the most specific and commonly used diagnostic parameters remain unclear. While recent discoveries of recurrent gene mutations have expanded our understanding, their prognostic significance is still being determined. A combination of advanced diagnostic techniques, including cytogenetics, immunophenotyping and molecular genetics such as qPCR, is essential for accurate diagnosis and disease management. The complexity of leukemia requires specialized laboratory assessments. Prognosis in leukemia is influenced by multiple factors, including patient performance status, genetic profiles and treatment response. Emerging studies on miRNAs and molecular responses to treatments such as imatinib highlight their potential prognostic value. However, most existing risk indicators remain prognostic rather than predictive, and the development of universally applicable scoring systems remains a challenge. As deep sequencing and targeted treatments evolve, the complexity of prognostic assessments will likely increase, necessitating the continuous refinement of therapeutic strategies.

Supplementary Data

Availability of data and materials

Not applicable.

Authors' contributions

JL, YW and LL conceived and designed the study. JL, CD and YW collected data. JL, CD, LL analyzed and interpreted data. JL, YW and CD wrote the draft of the manuscript. LL critically revised the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors are thankful to Dr Noor Bahadar (School of Life Sciences, Northeast Normal University) for providing technical support.

Funding

The present study was supported by the Natural Science Foundation of Jilin Province (grant no. YDZJ202301ZYTS042).

References

1 

Bose P and Bandyopadhyay S: A comprehensive assessment and classification of acute lymphocytic leukemia. Math Comput Appl. 29:452024.

2 

Sekeres MA, Guyatt G, Abel G, Alibhai S, Altman JK, Buckstein R, Choe H, Desai P, Erba H, Hourigan CS, et al: American Society of Hematology 2020 guidelines for treating newly diagnosed acute myeloid leukemia in older adults. Blood Adv. 4:3528–3549. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Tirado HA, Balasundaram N, Laaouimir L, Erdem A and van Gastel N: Metabolic crosstalk between stromal and malignant cells in the bone marrow niche. Bone Rep. 18:1016692023. View Article : Google Scholar : PubMed/NCBI

4 

Kaszuba CM, Rodems BJ, Sharma S, Franco EI, Ashton JM, Calvi LM and Bajaj J: Identifying bone marrow microenvironmental populations in myelodysplastic Syndrome and acute myeloid leukemia. J Vis Exp. Nov 10–2023.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

5 

Ye B, Sheng Y, Zhang M, Hu Y and Huang H: Early detection and intervention of clonal hematopoiesis for preventing hematological malignancies. Cancer Lett. 538:2156912022. View Article : Google Scholar : PubMed/NCBI

6 

Coombs CC, Zehir A, Devlin S, Zehir A, Devlin S, Kishtagari A, Hyman DM, Solit D, Robson M, Baselga J, et al: Mutational analysis of clonal hematopoiesis in solid tumor patients Illustrates the critical role of systemic anti-cancer therapies in the evolution of somatic leukemia disease alleles. Blood. 128:372016. View Article : Google Scholar

7 

Stengel A, Baer C, Walter W, Meggendorfer M, Kern W, Haferlach T and Haferlach C: Mutational patterns and their correlation to CHIP-related mutations and age in hematological malignancies. Blood Adv. 5:4426–4434. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Shiozawa Y, Havens AM, Pienta KJ and Taichman RS: The bone marrow niche: Habitat to hematopoietic and mesenchymal stem cells, and unwitting host to molecular parasites. Leukemia. 22:941–950. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Chowdhury MM: Stem cell in cancer disease cure vs cause. 2022, https://dspace.bracu.ac.bd/xmlui/bitstream/handle/10361/17759/18146041_PHR.pdf?sequence=1.

10 

Hoffman R: Progress in the development of systems for in vitro expansion of human hematopoietic stem cells. Curr Opin Hematol. 6:184–191. 1999. View Article : Google Scholar : PubMed/NCBI

11 

Landau DA, Carter SL, Getz G and Wu CJ: Clonal evolution in hematological malignancies and therapeutic implications. Leukemia. 28:34–43. 2014. View Article : Google Scholar

12 

Wang X, Huang S and Chen JL: Understanding of leukemic stem cells and their clinical implications. Mol Cancer. 16:22017. View Article : Google Scholar : PubMed/NCBI

13 

Komarova NL, Burger JA and Wodarz D: Evolution of ibrutinib resistance in chronic lymphocytic leukemia (CLL). Proc Natl Acad Sci USA. 111:13906–13911. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Shimony S, Stahl M and Stone RM: Acute myeloid leukemia: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol. 98:502–526. 2023. View Article : Google Scholar : PubMed/NCBI

15 

Jabbour E and Kantarjian H: Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring. Am J Hematol. 93:442–459. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Miranda-Filho A, Piñeros M, Ferlay J, Soerjomataram I, Monnereau A and Bray F: Epidemiological patterns of leukaemia in 184 countries: A population-based study. Lancet Haematol. 5:e14–e24. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Lin K, Jia H, Cao M, Xu T, Chen Z, Song X, Miao Y, Yao T, Dong C, Shao J, et al: Epidemiological characteristics of leukemia in China, 2005-2017: A log-linear regression and age-period-cohort analysis. BMC Public Health. 23:16472023. View Article : Google Scholar : PubMed/NCBI

18 

Zeidan AM, Mahmoud D, Kucmin-Bemelmans IT, Alleman CJ, Hensen M, Skikne B and Smith BD: Economic burden associated with acute myeloid leukemia treatment. Expert Rev Hematol. 9:79–89. 2016. View Article : Google Scholar

19 

Arber DA, Campo E and Jaffe ES: Advances in the classification of myeloid and lymphoid neoplasms. Virchows Arch. 482:1–9. 2023. View Article : Google Scholar : PubMed/NCBI

20 

Al-Bayati A, Al-Bayti A and Husain V: A short review about chronic myeloid leukemia. J Life Bio Sci Res. 4:15–19. 2023. View Article : Google Scholar

21 

Sell S: Leukemia: Stem cells, maturation arrest, and differentiation therapy. Stem Cell Rev. 1:197–206. 2005. View Article : Google Scholar

22 

Cheng Y, Luo H and Kharas MG: Rubbing out leukemia stem cells by erasing the eraser. Cell Stem Cell. 27:3–5. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Kassahun W, Tesfaye G, Bimerew LG, Fufa D, Adissu W and Yemane T: Prevalence of Leukemia and Associated Factors among Patients with Abnormal Hematological Parameters in Jimma Medical Center, Southwest Ethiopia: A Cross-Sectional Study. Adv Hematol. 2020:1–7. 2020. View Article : Google Scholar

24 

Jahic A, Iljazovic E, Hasic S, Arnautovic AC, Sabitovic D, Mesanovic S, Sahovic H and Simendic V: Prognostic Parameters of Acute Myeloid Leukaemia at Presentation. Med Arch. 71:20–24. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Zhang H, Luo XQ, Zhang P, Huang LB, Zheng YS, Wu J, Zhou H, Qu LH, Xu L and Chen YQ: MicroRNA patterns associated with clinical prognostic parameters and CNS relapse prediction in pediatric acute leukemia. PLoS One. 4:e78262009. View Article : Google Scholar : PubMed/NCBI

26 

Kardum-Skelin I, Planinc-Peraica A, Ostojić Kolonić S, Radić-Kristo D, Milas M, Vrhovac R, Sustercić D, Minigo H and Jaksić B: Clinical and laboratory prognostic parameters for leukemic types of chronic lymphoproliferative diseases. Acta Med Croatica. 62:351–364. 2008.In Croatian.

27 

Othieno-Abinya NA, Mwanda WO, Maina MD, Odhiambo AO, Mwanzi SA and Oyiro PO: Haematological parameters in chronic myeloid leukaemia as determinants of clinical manifestations in this disease. J Clin Oncol. 34:e185292016. View Article : Google Scholar

28 

Spiers AS: Management of the chronic leukemias: Special considerations in the elderly patient. part II: Chronic granulocytic leukemia. Hematology. 6:357–380. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Sharma K, Puniyani RR, Bhat SV, Advani SH, Hegde U and Rao S: Blood viscosity parameter correlation with types of leukemia. Physiol Chem Phys Med NMR. 24:159–164. 1992.PubMed/NCBI

30 

Germing U, Strupp C, Aivado M and Gattermann N: New prognostic parameters for chronic myelomonocytic leukemia? Blood. 100:731–732; author reply 732-3. 2002. View Article : Google Scholar

31 

Tonorezos ES, Robien K, Eshelman-Kent D, Moskowitz CS, Church TS, Ross R and Oeffinger KC: Contribution of diet and physical activity to metabolic parameters among survivors of childhood leukemia. Cancer Causes Control. 24:313–321. 2013. View Article : Google Scholar :

32 

Haferlach C, Dicker F, Weiss T, Schnittger S, Beck C, Grote-Metke A, Oruzio D, Kern W and Haferlach T: Toward a comprehensive prognostic scoring system in chronic lymphocytic leukemia based on a combination of genetic parameters. Genes Chromosomes Cancer. 49:851–859. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Jaeger BA, Tauer JT, Ulmer A, Kuhlisch E, Roth HJ and Suttorp M: Changes in bone metabolic parameters in children with chronic myeloid leukemia on imatinib treatment. Med Sci Monit. 18:CR721–CR728. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Letestu R, Lévy V, Eclache V, Baran-Marszak F, Vaur D, Naguib D, Schischmanoff O, Katsahian S, Nguyen-Khac F, Davi F, et al: Prognosis of Binet stage A chronic lymphocytic leukemia patients: the strength of routine parameters. Blood. 116:4588–4590. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Negrini M, Cutrona G, Bassi C, Fabris S, Zagatti B, Colombo M, Ferracin M, D'Abundo L, Saccenti E, Matis S, et al: microRNA ome expression in chronic lymphocytic leukemia: Comparison with Normal B-cell subsets and correlations with prognostic and clinical parameters. Clin Cancer Res. 20:4141–4153. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Rigolin GM, Cibien F, Martinelli S, Formigaro L, Rizzotto L, Tammiso E, Saccenti E, Bardi A, Cavazzini F, Ciccone M, et al: Chromosome aberrations detected by conventional karyotyping using novel mitogens in chronic lymphocytic leukemia with 'normal' FISH: Correlations with clinicobiologic parameters. Blood. 119:2310–2313. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Babusikova O, Stevulova L and Fajtova M: Immunophenotyping parameters as prognostic factors in T-acute leukemia patients. Neoplasma. 56:508–513. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Joncourt F, Oberlischraummli AE, Stadler M, Buser K, Franscini L, Fey MF and Cerny T: Patterns of Drug Resistance Parameters in Adult Leukemia. Leuk Lymphoma. 17:101–109. 1995. View Article : Google Scholar : PubMed/NCBI

39 

Baumgartner A, Zueger N, Bargetzi A, Medinger M, Passweg JR, Stanga Z, Mueller B, Bargetzi M and Schuetz P: Association of nutritional parameters with clinical outcomes in patients with acute myeloid leukemia undergoing haematopoietic stem cell transplantation. Ann Nutr Metab. 69:89–98. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Sillaber C, Mayerhofer M, Agis H, Sagaster V, Mannhalter C, Sperr WR, Geissler K and Valent P: Chronic myeloid leukemia: Pathophysiology, diagnostic parameters, and current treatment concepts. Wien Klin Wochenschr. 115:485–504. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Zittoun R, Cadiou M, Bayle C, Suciu S, Solbu G and Hayat M: Prognostic value of cytologic parameters in acute myelogenous leukemia. Cancer. 53:1526–1532. 1984. View Article : Google Scholar : PubMed/NCBI

42 

Piard C, Bressolle F, Fakhoury M, Zhang D, Yacouben K, Rieutord A and Jacqz-Aigrain E: A limited sampling strategy to estimate individual pharmacokinetic parameters of methotrexate in children with acute lymphoblastic leukemia. Cancer Chemother Pharmacol. 60:609–620. 2006. View Article : Google Scholar

43 

Halonen P, Mattila J, Suominen P, Ruuska T, Salo MK and Mäkipernaa A: Iron overload in children who are treated for acute lymphoblastic leukemia estimated by liver siderosis and serum iron parameters. Pediatrics. 111:91–96. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Prabhu M, Kochupillai V, Sharma S, Ramachandran P, Sundaram KR, Bijlani L and Ahuja RK: Prognostic assessment of various parameters in chronic myeloid leukemia. Cancer. 58:1357–1360. 1986. View Article : Google Scholar : PubMed/NCBI

45 

Shinawi T, Hill V, Dagklis A, Baliakas P, Stamatopoulos K, Agathanggelou A, Stankovic T, Maher ER, Ghia P and Latif F: KIBRAgene methylation is associated with unfavorable biological prognostic parameters in chronic lymphocytic leukemia. Epigenetics. 7:211–215. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Royer-Pokora B, Beug H, Claviez M, Winkhardt HJ, Friis RR and Graf T: Transformation parameters in chicken fibroblasts transformed by AEV and MC29 avian leukemia viruses. Cell. 13:751–760. 1978. View Article : Google Scholar : PubMed/NCBI

47 

Oliveira GB, Pereira FG, Metze K and Lorand-Metze I: Spontaneous apoptosis in chronic lymphocytic leukemia and its relationship to clinical and cell kinetic parameters. Cytometry. 46:329–335. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Hofmann-Lehmann R, Holznagel E, Ossent P and Lutz H: Parameters of disease progression in long-term experimental feline retrovirus (feline immunodeficiency virus and feline leukemia virus) infections: hematology, clinical chemistry, and lymphocyte subsets. Clin Diagn Lab Immunol. 4:33–42. 1997. View Article : Google Scholar : PubMed/NCBI

49 

Chiaretti S, Marinelli M, Del Giudice I, Bonina S, Piciocchi A, Messina M, Vignetti M, Rossi D, Di Maio V, Mauro FR, et al: NOTCH1, SF3B1, BIRC3andTP53mutations in patients with chronic lymphocytic leukemia undergoing first-line treatment: correlation with biological parameters and response to treatment. Leuk Lymphoma. 55:2785–2792. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Li J, Zheng R, Niu J, Song X, Wu W, Fan R and Gong T: Correlation of intravoxel incoherent motion parameters and histological characteristics from infiltrated marrow in patients with acute leukemia. J Magn Reson Imaging. 51:1720–1726. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Dhasmana R, Prakash A, Gupta N and Verma SK: Ocular manifestations in leukemia and myeloproliferative disorders and their association with hematological parameters. Ann Afr Med. 15:97–103. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Omer SH, AA K, IM O, OH M, HN A and Elamin BK: Prevalence of the different FAB sub type of Acute Myeloid Leukemia related to hematological parameters in Sudanese. J Hematol Blood Disord. 3:1022017. View Article : Google Scholar

53 

Heintel D, Schwarzinger I, Chizzali-Bonfadin C, Thalhammer R, Schwarzmeier J, Fritzer-Szekeres M, Weltermann A, Simonitsch I, Lechner K and Jaeger U: Association of CD38 antigen expression with other prognostic parameters in early stages of chronic lymphocytic leukemia. Leuk Lymphoma. 42:1315–1321. 2001. View Article : Google Scholar

54 

Molica S, Cutrona G, Vitelli G, Mirabelli R, Molica M, Digiesi G, Ribatti D, Ferrarini M and Vacca A: Markers of increased angiogenesis and their correlation with biological parameters identifying high-risk patients in early B-cell chronic lymphocytic leukemia. Leuk Res. 31:1575–1578. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Levine PH, Merrill DA, Bethlenfalvay NC, Dabich L, Stevens DA and Waggoner DE: A longitudinal comparison of antibodies to epstein-barr virus and clinical parameters in chronic lymphocytic leukemia and chronic myelocytic leukemia. Blood. 38:479–484. 1971. View Article : Google Scholar : PubMed/NCBI

56 

Martell RE, Peterson BL, Cohen HJ, Petros WP, Rai KR, Morrison VA, Elias L, Shepherd L, Hines J, Larson RA, et al: Analysis of age, estimated creatinine clearance and pretreatment hematologic parameters as predictors of fludarabine toxicity in patients treated for chronic lymphocytic leukemia: A CALGB (9011) coordinated intergroup study. Cancer Chemother Pharmacol. 50:37–45. 2002. View Article : Google Scholar : PubMed/NCBI

57 

Bergmann MA, Eichhorst BF, Busch R, Adorf D, Stilgenbauer S, Eckart MJ, Wendtner CM and Hallek M; GCLLSG: Prospective Evaluation of Prognostic Parameters in Early Stage Chronic Lymphocytic Leukemia (CLL): Results of the CLL1-Protocol of the German CLL Study Group (GCLLSG). Blood. 110:6252007. View Article : Google Scholar

58 

Sinisalo M, Aittoniemi J, Koski T, Tobin G, Thunberg U, Sundström C, Rosenquist R, Käyhty H and Vilpo J: Similar Humoral Immunity Parameters in Chronic Lymphocytic Leukemia Patients Independent of VH Gene Mutation Status. Leuk Lymphoma. 45:2451–2454. 2004. View Article : Google Scholar : PubMed/NCBI

59 

Browman G, Goldberg J, Gottlieb AJ, Preisler HD, Azarnia N, Priore RL, Brennan JK, Vogler WR, Winton EF, Miller KB, et al: The clonogenic assay as a reproducible in vitro system to study predictive parameters of treatment outcome in acute nonlymphoblastic leukemia. Am J Hematol. 15:227–235. 1983. View Article : Google Scholar : PubMed/NCBI

60 

Wieser R, Scheideler M, Hackl H, Engelmann M, Schneckenleithner C, Hiden K, Papak C, Trajanoski Z, Sill H and Fonatsch C: microRNAs in acute myeloid leukemia: Expression patterns, correlations with genetic and clinical parameters, and prognostic significance. Genes Chromosomes Cancer. 49:193–203. 2010. View Article : Google Scholar

61 

Valet G, Repp R, Link H, Ehninger A and Gramatzki MM; SHG-AML study group: Pretherapeutic identification of high-risk acute myeloid leukemia (AML) patients from immunophenotypic, cytogenetic, and clinical parameters. Cytometry B Clin Cytom. 53:4–10. 2003. View Article : Google Scholar : PubMed/NCBI

62 

Valent P, Agis H, Sperr W, Sillaber C and Horny HP: Diagnostic and prognostic value of new biochemical and immunohistochemical parameters in chronic myeloid leukemia. Leuk Lymphoma. 49:635–638. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Zimm S, Reaman G, Murphy RF and Poplack DG: Biochemical parameters of mercaptopurine activity in patients with acute lymphoblastic leukemia. Cancer Res. 46:1495–1498. 1986.PubMed/NCBI

64 

Yang JH, Kim Y, Lim J, Kim M, Oh EJ, Lee HK, Park YJ, Min WS, Cho B, Lee K and Han K: Determination of acute leukemia lineage with new morphologic parameters available in the complete blood cell count. Ann Clin Lab Sci. 44:19–26. 2014.PubMed/NCBI

65 

Olson KC, Moosic KB, Jones MK, Larkin PMK, Olson TL, Toro MF, Fox TE, Feith DJ and Loughran TP Jr: Large granular lymphocyte leukemia serum and corresponding hematological parameters reveal unique cytokine and sphingolipid biomarkers and associations with STAT3 mutations. Cancer Med. 9:6533–6549. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Srinivasan S, Cui H, Gao Z, Liu M, Lu S, Mkandawire W, Narykov O, Sun M and Korkin D: Structural Genomics of SARS-CoV-2 indicates evolutionary conserved functional regions of viral proteins. Viruses. 12:3602020. View Article : Google Scholar : PubMed/NCBI

67 

Hallek M: Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment. Am J Hematol. 94:1266–1287. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Feller N, van der Pol MA, van Stijn A, Weijers GW, Westra AH, Evertse BW, Ossenkoppele GJ and Schuurhuis GJ: MRD parameters using immunophenotypic detection methods are highly reliable in predicting survival in acute myeloid leukaemia. Leukemia. 18:1380–1390. 2004. View Article : Google Scholar : PubMed/NCBI

69 

Mohammadi Kanesbi M, Jarahi L and Keramati MR: Blood parameters in treatment with arsenic trioxide in acute promyelocytic leukemia: A systematic review. Int J Hematol Oncol Stem Cell Res. 14:130–139. 2020.PubMed/NCBI

70 

Al-Bayaa IM, Al-Rubaie HA and Al-Shammari HH: Evaluation of hepatocyte growth factor in iraqi patients with acute myeloid leukemia: Its correlation with clinical parameters and response to induction therapy. Open Access Maced J Med Sci. 8:49–53. 2020. View Article : Google Scholar

71 

Khazaal MS, Hamdan FB and Al-Mayah QS: Association of BCR/ABL transcript variants with different blood parameters and demographic features in Iraqi chronic myeloid leukemia patients. Mol Genet Genomic Med. 7:e8092019. View Article : Google Scholar : PubMed/NCBI

72 

Ogasawara A, Matsushita H, Tanaka Y, Shirasugi Y, Ando K, Asai S and Miyachi H: A simple screening method for the diagnosis of chronic myeloid leukemia using the parameters of a complete blood count and differentials. Clin Chim Acta. 489:249–253. 2019. View Article : Google Scholar

73 

Michailov Y, Lunenfeld E, Kapilushnik J, Friedler S, Meese E and Huleihel M: Acute myeloid leukemia affects mouse sperm parameters, spontaneous acrosome reaction, and fertility capacity. Int J Mol Sci. 20:2192019. View Article : Google Scholar : PubMed/NCBI

74 

Virk H, Varma N, Naseem S, Bihana I and Sukhachev D: Utility of cell population data (VCS parameters) as a rapid screening tool for Acute Myeloid Leukemia (AML) in resource-constrained laboratories. J Clin Lab Anal. 33:e226792019. View Article : Google Scholar

75 

Vigón L, Luna A, Galán M, Rodríguez-Mora S, Fuertes D, Mateos E, Piris-Villaespesa M, Bautista G, San José E, Rivera-Torres J, et al: Identification of immunological parameters as predictive biomarkers of relapse in patients with chronic myeloid leukemia on treatment-free remission. J Clin Med. 10:422020. View Article : Google Scholar : PubMed/NCBI

76 

Mahmood R, Khan SA, Altaf C, Malik HS and Khadim MT: Clinicohematological parameters and outcomes in a cohort of chronic lymphocytic leukemia patients with Deletion 17p from Pakistan. Blood Res. 53:276–280. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Kim H, Ahn SI, Jhoo JW and Kim GY: Comparison of Allergic Parameters between Whey Protein Concentrate and Its Hydrolysate in Rat Basophilic Leukemia (RBL)-2H3 Cells. Korean J Food Sci Anim Resour. 38:780–793. 2018.PubMed/NCBI

78 

Schillinger F, Sourdeau E, Boubaya M, Baseggio L, Clauser S, Cornet E, Debord C, Defour JP, Dubois F, Eveillard M, et al: A new approach for diagnosing chronic myelomonocytic leukemia using structural parameters of Sysmex XN™ analyzers in routine laboratory practice. Scand J Clin Lab Invest. 78:159–164. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Khan MI: Acute Myeloid Leukemia: Pattern of clinical and hematological parameters in a tertiary care centre. Int J Pathol. 11:58–63. 2018.

80 

Podszywalow-Bartnicka P, Kominek A, Wolczyk M, Kolba MD, Swatler J and Piwocka K: Characteristics of live parameters of the HS-5 human bone marrow stromal cell line cocultured with the leukemia cells in hypoxia, for the studies of leukemia-stroma cross-talk. Cytometry A. 93:929–940. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Nägele V, Kratzer A, Zugmaier G, Holland C, Hijazi Y, Topp MS, Gökbuget N, Baeuerle PA, Kufer P, Wolf A and Klinger M: Changes in clinical laboratory parameters and pharmacodynamic markers in response to blinatumomab treatment of patients with relapsed/refractory ALL. Exp Hematol Oncol. 6:142017. View Article : Google Scholar : PubMed/NCBI

82 

Lekovic D, Gotic M, Milic N, Zivojinovic B, Jovanovic J, Colovic N, Milosevic V and Bogdanovic A: Predictive parameters for imatinib failure in patients with chronic myeloid leukemia. Hematology. 22:460–466. 2017.PubMed/NCBI

83 

Burhan I: Estimation Of Alpha-Fetoprotein (AFP) and some of biochemical parameters in leukemia patients. World J Pharm Pharma Sci. 5:2275–2283. 2017.

84 

Elzubeir MA, Angi A, Rahoum H, Ismail IA, Mohammed ME, Yousef MM, Khair SM, Namarq Alaaldeen SA, Osman RAM, Duaa Ali AA, et al: Prognostic Significance of Immune Function Parameters (CD4, CD8 and CD4/CD8 ratio) in Sudanese Patients with Chronic Lymphocytic leukemia. Int J Multidiscip Curr Res. 4:7650–653. 2016.

85 

Wenn K, Tomala L, Wilop S, Vankann L, Hasenbank C, Frank O, Hochhaus A, Giles FJ, Lange T, Müller MC, et al: Telomere length at diagnosis of chronic phase chronic myeloid leukemia (CML-CP) identifies a subgroup with favourable prognostic parameters and molecular response according to the ELN criteria after 12 months of treatment with nilotinib. Leukemia. 29:2402–2404. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Trendowski M, Wong V, Zoino JN, Christen TD, Gadeberg L, Sansky M and Fondy TP: Preferential enlargement of leukemia cells using cytoskeletal-directed agents and cell cycle growth control parameters to induce sensitivity to low frequency ultrasound. Cancer Lett. 360:160–170. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Huang Z, Liu WJ, Guo QL and Liu CY: Platelet parameters and expression of platelet membrane glycoprotein in childhood acute lymphoblastic leukemia. Genet Mol Res. 14:16074–16089. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Rabizadeh E, Pickholtz I, Barak M, Isakov E, Zimra Y and Froom P: Acute leukemia detection rate by automated blood count parameters and peripheral smear review. Int J Lab Hematol. 37:44–49. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Koltan S, Debski R, Koltan A, Grzesk E, Tejza B, Eljaszewicz A, Gackowska L, Kubicka M, Kolodziej B, Kurylo-Rafinska B, et al: Phenotype of NK cells determined on the basis of selected immunological parameters in children treated due to acute lymphoblastic leukemia. Medicine (Baltimore). 94:e23692015. View Article : Google Scholar : PubMed/NCBI

90 

Aljamaan K, Aljumah TK, Aloraibi S, Absar M and Iqbal Z: Low Frequency of ETV6-RUNX1 (t 12; 21) in Saudi Arabian pediatric acute lymphoblastic leukemia patients: Association with clinical parameters and early remission. Asian Pac J Cancer Prev. 16:7523–7527. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Rafiq N, Iqbal T, Shahid M and Muhammad F: Hematological and Biochemical Parameters in Pakistani Chronic Lymphoblastic Leukemia Patients. Pak J Life Soc Sci. 12:16–19. 2014.

92 

Pietras W, Chaber R, Pela H, Trybucka K and Chybicka A: The recovery of immune system parameters in children following lymphoblastic leukemia therapy-preliminary report. Adv Clin Exp Med. 23:97–102. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Deschler B, Ihorst G, Platzbecker U, Germing U, März E, de Figuerido M, Fritzsche K, Haas P, Salih HR, Giagounidis A, et al: Parameters detected by geriatric and quality of life assessment in 195 older patients with myelodysplastic syndromes and acute myeloid leukemia are highly predictive for outcome. Haematologica. 98:208–216. 2013. View Article : Google Scholar :

94 

Mauro MJ, Cortes JE, Kim DW, Pinilla-Ibarz J, Coutre PI, Paquette R, Chuah C, Nicolini FE, Apperley J, Khoury HJ, et al: Multivariate Analyses of the Clinical and Molecular Parameters Associated with Efficacy and Safety in Patients with Chronic Myeloid Leukemia (CML) and Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia (Ph+ ALL) Treated with Ponatinib in the PACE Trial. Blood. 120:37472012. View Article : Google Scholar

95 

Dong HJ, Miao KR, Qiao C, Zhuang Y, Shen WY, Hong M, Fan L, Liu P, Xu W and Li JY: Polymorphisms and haplotypes in multidrug resistance 1 gene are not associated with chronic lymphocytic leukemia susceptibility and prognostic parameters of chronic lymphocytic leukemia in Chinese population. Leuk Lymphoma. 52:1003–1009. 2011. View Article : Google Scholar : PubMed/NCBI

96 

Onida F, Kantarjian HM, Smith TL, Ball G, Keating MJ, Estey EH, Glassman AB, Albitar M, Kwari MI and Beran M: Prognostic factors and scoring systems in chronic myelomonocytic leukemia: A retrospective analysis of 213 patients. Blood. 99:840–849. 2002. View Article : Google Scholar : PubMed/NCBI

97 

Sagatys EM and Zhang L: Clinical and laboratory prognostic indicators in chronic lymphocytic leukemia. Cancer Control. 19:18–25. 2012. View Article : Google Scholar

98 

Jinlong S, Lin F, Yonghui L, Li Y and Weidong W: Identification of let-7a-2-3p or/and miR-188-5p as prognostic biomarkers in cytogenetically normal acute myeloid leukemia. PLoS One. 10:e01180992015. View Article : Google Scholar : PubMed/NCBI

99 

Marcucci G, Mrózek K, Radmacher MD, Garzon R and Bloomfield CD: The prognostic and functional role of microRNAs in acute myeloid leukemia. Blood. 117:1121–1129. 2011. View Article : Google Scholar :

100 

Labib HA, Elantouny NG, Ibrahim NF and Alnagar AA: Upregulation of microRNA-21 is a poor prognostic marker in patients with childhood B cell acute lymphoblastic leukemia. Hematology. 22:392–397. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Neaga A, Bagacean C, Tempescul A, Jimbu L, Mesaros O, Blag C, Tomuleasa C, Bocsan C, Gaman M and Zdrenghea M: MicroRNAs associated with a good prognosis of acute myeloid leukemia and their effect on macrophage polarization. Front Immunol. 11:5829152021. View Article : Google Scholar : PubMed/NCBI

102 

Ciesielska M, Orzechowska B, Gamian A and Kazanowska B: Epidemiology of childhood acute leukemias. Postep Hig Med Dosw. 78:22–36. 2024.In Polish. View Article : Google Scholar

103 

Mari JF, de Miranda ÉJFP, Mendes-Correa MC, Chow FC and Vidal JE: Progressive multifocal leukoencephalopathy and spectrum of predisposing conditions: A 20-year retrospective cohort study in a tertiary center in São Paulo, Brazil. Neurol Sci. Jul 15–2024.Epub ahead of print. View Article : Google Scholar

104 

Aureli A, Marziani B, Venditti A, Sconocchia T and Sconocchia G: Acute Lymphoblastic Leukemia Immunotherapy Treatment: Now, Next, and Beyond. Cancers (Basel). 15:33462023. View Article : Google Scholar : PubMed/NCBI

105 

Giannakaki AG, Giannakaki MN, Oikonomou E, Nikolettos K, Bothou A, Kotanidou S, Andreou S, Kritsotaki N, Nalmpanti T, Spanoudakis E, et al: Leukemia in pregnancy: Diagnosis and therapeutic approach (Review). Mol Clin Oncol. 21:792024. View Article : Google Scholar : PubMed/NCBI

106 

van der Straten L, Levin MD, Dinnessen MAW, Visser O, Posthuma EFM, Doorduijn JK, Langerak AW, Kater AP and Dinmohamed AG: Risk of second primary malignancies in patients with chronic lymphocytic leukemia: A population-based study in the Netherlands, 1989-2019. Blood Cancer J. 13:152023. View Article : Google Scholar : PubMed/NCBI

107 

Jain M, Rai A, Kushwaha R, Yadav G, Mishra S, Verma SP, Singh US, Tripathi AK and Kumar A: A cross sectional study on T Cells and Subsets in Chronic Lymphocytic Leukemia and their Association with Clinical Phenotype. Indian J Hematol Blood Transfus. 37:563–568. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Qurat Ul Ain, Akbar S, Gull S, Hussain M and Ayesha N: Leukemia detection using machine and deep learning through microscopic images-A review. Studies in Big Data. Jul 7–2022.Epub ahead of print.

109 

Alcazer V, Le Meur G, Roccon M, Barriere S, Le Calvez B, Badaoui B, Spaeth A, Kosmider O, Freynet N, Eveillard M, et al: Evaluation of a machine-learning model based on laboratory parameters for the prediction of acute leukaemia subtypes: A multicentre model development and validation study in France. Lancet Digit Health. 6:e323–e333. 2024. View Article : Google Scholar : PubMed/NCBI

110 

Vineela M, Reddy GDS, Karthik G, Muthukumaran N and Al Deen SH: Classification of Leukemia White Blood Cell Cancer. In: 2024 International Conference on Inventive Computation Technologies (ICICT); pp. 1233–1236. 2024

111 

Papadakis S, Liapis I, Papadhimitriou SI, Spanoudakis E, Kotsianidis I and Liapis K: Approach to acute myeloid leukemia with increased eosinophils and basophils. J Clin Med. 13:8762024. View Article : Google Scholar : PubMed/NCBI

112 

Ahmed MR and Anis M: In vitro regeneration and the antioxidant enzymatic system on acclimatization of micropropagated Vitex trifolia L. Agroforest Syst. 88:437–447. 2014. View Article : Google Scholar

113 

Li H, Xu C, Xin B, Zheng C, Zhao Y, Hao K, Wang Q, Wahl RL, Wang X and Zhou Y: 18F-FDG PET/CT radiomic analysis with machine learning for identifying bone marrow involvement in the patients with suspected relapsed acute leukemia. Theranostics. 9:4730–4739. 2019. View Article : Google Scholar :

114 

Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, Dombret H, Ebert BL, Fenaux P, Larson RA, et al: Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 129:424–447. 2017. View Article : Google Scholar :

115 

Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H and Thiele J: WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. WHO Classification of Tumours; 2017

116 

Tallman MS, Wang ES, Altman JK, Appelbaum FR, Bhatt VR, Bixby D, Coutre SE, De Lima M, Fathi AT, Fiorella M, et al: Acute Myeloid Leukemia, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 17:721–749. 2019. View Article : Google Scholar : PubMed/NCBI

117 

Jondreville L, Krzisch D, Chapiro E and Nguyen-Khac F: The complex karyotype and chronic lymphocytic leukemia: prognostic value and diagnostic recommendations. Am J Hematol. 95:1361–1367. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Andreieva SV, Korets KV, Tsyapka OM and Skorokhod IM: Comparative Characteristics of Abnormal Karyotypes in Secondary Acute Leukemia. Cytol Genet. 58:292–299. 2024. View Article : Google Scholar

119 

Olde Nordkamp L, Mellink C, van der Schoot E and van den Berg H: Karyotyping FISH, and PCR in acute lymphoblastic leukemia: competing or complementary diagnostics? J Pediatr Hematol Oncol. 31:930–935. 2009. View Article : Google Scholar : PubMed/NCBI

120 

Camuset M, Le Calvez B, Theisen O, Godon C, Grain A, Thomas C, Couec ML, Béné MC, Rialland F and Eveillard M: Added value of molecular karyotype in childhood acute lymphoblastic leukemia. Cancer Innov. 2:513–523. 2023. View Article : Google Scholar : PubMed/NCBI

121 

Kibler CE and Chabot-Richards DS: Molecular Pathology of Leukemia. Molecular Surgical Pathology. Cheng L, Netto GJ and Eble JN: Springer International Publishing; Cham: pp. 681–709. 2023, View Article : Google Scholar

122 

Bayani J and Squire JA: Preparation of cytogenetic specimens from tissue samples. Curr Protoc Cell Biol. Chapter 22: Unit 22.2. 2004. View Article : Google Scholar

123 

Stölzel F, Mohr B, Kramer M, Oelschlägel U, Bochtler T, Berdel WE, Kaufmann M, Baldus CD, Schäfer-Eckart K, Stuhlmann R, et al: Karyotype complexity and prognosis in acute myeloid leukemia. Blood Cancer J. 6:e3862016. View Article : Google Scholar : PubMed/NCBI

124 

Kiefer Y, Schulte C, Tiemann M and Bullerdiek J: Chronic lymphocytic leukemia-associated chromosomal abnormalities and miRNA deregulation. Appl Clin Genet. 5:21–28. 2012.PubMed/NCBI

125 

Alhourani E, Rincic M, Othman MA, Pohle B, Schlie C, Glaser A and Liehr T: Comprehensive chronic lymphocytic leukemia diagnostics by combined multiplex ligation dependent probe amplification (MLPA) and interphase fluorescence in situ hybridization (iFISH). Mol Cytogenet. 7:792014. View Article : Google Scholar : PubMed/NCBI

126 

Han X, Jin C, Zheng G, Li Y, Wang Y, Zhang E, Zhu H and Cai Z: Acute myeloid leukemia with CPSF6-RARG fusion resembling acute promyelocytic leukemia with extramedullary infiltration. Ther Adv Hematol. 12:20406207209769842021. View Article : Google Scholar : PubMed/NCBI

127 

Gupta V, Shariff M, Bajwa R, Patel I, Ayyad HA, Levitt MJ, Mencel PJ and Hossain MA: Acute myeloid leukemia acquiring promyelocytic leukemia-retinoic acid receptor alpha at relapse. World J Oncol. 10:153–156. 2019. View Article : Google Scholar : PubMed/NCBI

128 

Hochhaus A, Baccarani M, Silver RT, Schiffer C, Apperley JF, Cervantes F, Clark RE, Cortes JE, Deininger MW, Guilhot F, et al: European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia. 34:966–984. 2020. View Article : Google Scholar : PubMed/NCBI

129 

Danylesko I, Jacoby E, Yerushalmi R, Shem-Tov N, Besser MJ, Vernitsky H, Marcu-Malina V, Shimoni A, Avigdor A and Nagler A: Remission of acute myeloid leukemia with t(8;21) following CD19 CAR T-cells. Leukemia. 34:1939–1942. 2020. View Article : Google Scholar : PubMed/NCBI

130 

Bellesi S, Schiaffini G, Contegiacomo A, Maiolo E, Iacovelli C, Malafronte R, D'Innocenzo S, Alma E, Bellisario F, Viscovo M, et al: Enhancing lymphoma diagnosis on core needle biopsies: Integrating immunohistochemistry with flow cytometry. Cytometry B Clin Cytom. Jun 14–2024.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

131 

Kurosawa S and Nakazato T: Immunohistochemical differentiation of extramedullary acute myeloid leukemia from blastic plasmacytoid dendritic cell neoplasm. Int J Hematol. 120:391–393. 2024. View Article : Google Scholar : PubMed/NCBI

132 

Hisada Y, Archibald SJ, Bansal K, Chen Y, Dai C, Dwarampudi S, Balas N, Hageman L, Key NS, Bhatia S, et al: Biomarkers of bleeding and venous thromboembolism in patients with acute leukemia. J Thromb Haemost. 22:1984–1996. 2024. View Article : Google Scholar : PubMed/NCBI

133 

De Coninck S, De Smedt R, Lintermans B, Reunes L, Kosasih HJ, Reekmans A, Brown LM, Van Roy N, Palhais B, Roels J, et al: Targeting hyperactive platelet-derived growth factor receptor-β signaling in T-cell acute lymphoblastic leukemia and lymphoma. Haematologica. 109:1373–1384. 2024.

134 

Elliott MA and Tefferi A: Chronic neutrophilic leukemia: 2018 update on diagnosis, molecular genetics and management. Am J Hematol. 93:578–587. 2018. View Article : Google Scholar : PubMed/NCBI

135 

Fiala GJ and Minguet S: Caveolin-1: The Unnoticed Player in TCR and BCR Signaling. Adv Immunol. 137:83–133. 2018. View Article : Google Scholar : PubMed/NCBI

136 

Apostolopoulos A, Symeonidis A and Zoumbos N: Prognostic significance of immune function parameters in patients with chronic lymphocytic leukaemia. Eur J Haematol. 44:39–44. 1990. View Article : Google Scholar : PubMed/NCBI

137 

Nardini G, Leopardi S and Bielli M: Clinical Hematology in Reptilian Species. Vet Clin North Am Exot Anim Pract. 16:1–30. 2013. View Article : Google Scholar : PubMed/NCBI

138 

Oliveira ALCSL, Zerillo L, Cruz LJ, Schomann T, Chan AB, de Carvalho TG, Souza SVP, Araújo AA, de Geus-Oei LF and de Araújo Júnior RF: Maximizing the potency of oxaliplatin coated nanoparticles with folic acid for modulating tumor progression in colorectal cancer. Mater Sci Eng C Mater Biol Appl. 120:1116782021. View Article : Google Scholar : PubMed/NCBI

139 

Belov L, Huang P, Barber N, Mulligan SP and Christopherson RI: Identification of repertoires of surface antigens on leukemias using an antibody microarray. Proteomics. 3:2147–2154. 2003. View Article : Google Scholar : PubMed/NCBI

140 

Hömig-Hölzel C and Savola S: Multiplex Ligation-dependent Probe Amplification (MLPA) in Tumor Diagnostics and Prognostics. Diagn Mol Pathol. 21:189–206. 2012. View Article : Google Scholar : PubMed/NCBI

141 

Ningombam A, Verma D, Kumar R, Singh J, Ali MS, Pandey AK, Singh I, Bakhshi S, Sharma A, Pushpam D, et al: Prognostic relevance of NPM1, CEBPA, and FLT3 mutations in cytogenetically normal adult AML patients. Am J Blood Res. 13:28–43. 2023.PubMed/NCBI

142 

Dietrich P, Gaza A, Wormser L, Fritz V, Hellerbrand C and Bosserhoff AK: Neuroblastoma RAS Viral Oncogene Homolog (NRAS) is a novel prognostic marker and contributes to sorafenib resistance in hepatocellular carcinoma. Neoplasia. 21:257–268. 2019. View Article : Google Scholar : PubMed/NCBI

143 

Li S, Mason C and Melnick A: Genetic and epigenetic heterogeneity in acute myeloid leukemia. Curr Opin Genet Dev. 36:100–106. 2016. View Article : Google Scholar : PubMed/NCBI

144 

Döhner H, Estey EH, Amadori S, Appelbaum FR, Büchner T, Burnett AK, Dombret H, Fenaux P, Grimwade D, Larson RA, et al: Diagnosis and management of acute myeloid leukemia in adults: Recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 115:453–474. 2010. View Article : Google Scholar

145 

Menssen AJ and Walter MJ: Genetics of progression from MDS to secondary leukemia. Blood. 136:50–60. 2020. View Article : Google Scholar : PubMed/NCBI

146 

Bond J, Touzart A, Leprêtre S, Graux C, Bargetzi M, Lhermitte L, Hypolite G, Leguay T, Hicheri Y, Guillerm G, et al: DNMT3A mutation is associated with increased age and adverse outcome in adult T-cell acute lymphoblastic leukemia. Haematologica. 104:1617–1625. 2019. View Article : Google Scholar : PubMed/NCBI

147 

Stone RM: The difficult problem of acute myeloid leukemia in the older adult. CA Cancer J Clin. 52:363–371. 2002. View Article : Google Scholar : PubMed/NCBI

148 

Appelbaum FR, Gundacker H, Head DR, Slovak ML, Willman CL, Godwin JE, Anderson JE and Petersdorf SH: Age and acute myeloid leukemia. Blood. 107:3481–3485. 2006. View Article : Google Scholar : PubMed/NCBI

149 

Manescu P, Narayanan P, Bendkowski C, Elmi M, Claveau R, Pawar V, Brown BJ, Shaw M, Rao A and Fernandez-Reyes D: Detection of acute promyelocytic leukemia in peripheral blood and bone marrow with annotation-free deep learning. Sci Rep. 13:25622023. View Article : Google Scholar : PubMed/NCBI

150 

Verma D, Yadav R, Ahirwar R and Meena P: Importance of bone marrow examination in cases of pancytopenia: A morphological study in a tertiary care center. Asian J Med Sci. 15:202–205. 2024. View Article : Google Scholar

151 

Rivera D, Cui W, Gao J, Peker D, Zhang QY, Dewar R, Qiu L, Konoplev S, Hu Z, Sasaki K, et al: Aleukemic chronic myeloid leukemia without neutrophilia and thrombocytosis: A Report From the BCR::ABL1 Pathology Group. Mod Pathol. 37:1004062024. View Article : Google Scholar

152 

Terwilliger T and Abdul-Hay M: Acute lymphoblastic leukemia: A comprehensive review and 2017 update. Blood Cancer J. 7:e5772017. View Article : Google Scholar : PubMed/NCBI

153 

Jabbour E and Kantarjian H: Chronic myeloid leukemia: 2020 update on diagnosis, therapy and monitoring. Am J Hematol. 95:691–709. 2020. View Article : Google Scholar : PubMed/NCBI

154 

Troussard X, Maître E and Cornet E: Hairy cell leukemia 2022: Update on diagnosis, risk-stratification, and treatment. Am J Hematol. 97:226–236. 2022. View Article : Google Scholar

155 

Wood ME: Chapter 57-Oncologic Emergencies (Including Hypercalcemia). Critical Care Secrets (Fifth Edition). Parsons PE and Wiener-Kronish JP: Mosby; Philadelphia, PA: pp. 404–409. 2013, View Article : Google Scholar

156 

Kang SH, Kim HK, Ham CK, Lee DS and Cho HI: Comparison of four hematology analyzers, CELL-DYN Sapphire, ADVIA 120, Coulter LH 750, and Sysmex XE-2100, in terms of clinical usefulness. Int J Lab Hematol. 30:480–486. 2008. View Article : Google Scholar : PubMed/NCBI

157 

He G, Jiang Z, Xue S, Sun X and Wang W: Expression of LDH and CEA in serum in the process of targeted therapy of lung adenocarcinoma and the association between them and prognosis. Oncol Lett. 17:4550–4556. 2019.PubMed/NCBI

158 

Oliveira LC, Romano LG, Prado-Junior BP, Covas DT, Rego EM and De Santis GC: Outcome of acute myeloid leukemia patients with hyperleukocytosis in Brazil. Med Oncol. 27:1254–1259. 2010. View Article : Google Scholar

159 

Zografos E, Zagouri F, Kalapanida D, Zakopoulou R, Kyriazoglou A, Apostolidou K, Gazouli M and Dimopoulos MA: Prognostic role of microRNAs in breast cancer: A systematic review. Oncotarget. 10:7156–7178. 2019. View Article : Google Scholar

160 

Davis-Dusenbery BN and Hata A: MicroRNA in Cancer: The Involvement of Aberrant MicroRNA Biogenesis Regulatory Pathways. Genes Cancer. 1:1100–1114. 2010. View Article : Google Scholar

161 

Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, Frankel WL, Morgan DL, Postier RG, Brackett DJ and Schmittgen TD: Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer. 120:1046–1054. 2007. View Article : Google Scholar

162 

Garzon R, Garofalo M, Martelli MP, Briesewitz R, Wang L, Fernandez-Cymering C, Volinia S, Liu CG, Schnittger S, Haferlach T, et al: Distinctive microRNA signature of acute myeloid leukemia bearing cytoplasmic mutated nucleophosmin. Proc Natl Acad Sci USA. 105:3945–3950. 2008. View Article : Google Scholar : PubMed/NCBI

163 

Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K, et al: Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 99:15524–15529. 2002. View Article : Google Scholar

164 

Mi S, Lu J, Sun M, Li Z, Zhang H, Neilly MB, Wang Y, Qian Z, Jin J, Zhang Y, et al: MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Proc Natl Acad Sci USA. 104:19971–19976. 2007. View Article : Google Scholar : PubMed/NCBI

165 

Marcucci G, Maharry K, Radmacher MD, Mrózek K, Vukosavljevic T, Paschka P, Whitman SP, Langer C, Baldus CD, Liu CG, et al: Prognostic significance of, and gene and microRNA expression signatures associated with, CEBPA mutations in cytogenetically normal acute myeloid leukemia with high-risk molecular features: A Cancer and Leukemia Group B Study. J Clin Oncol. 26:5078–5087. 2008. View Article : Google Scholar : PubMed/NCBI

166 

Li Z, Lu J, Sun M, Mi S, Zhang H, Luo RT, Chen P, Wang Y, Yan M, Qian Z, et al: Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proc Natl Acad Sci USA. 105:15535–15540. 2008. View Article : Google Scholar : PubMed/NCBI

167 

Abbott BL, Rubnitz JE, Tong X, Srivastava DK, Pui CH, Ribeiro RC and Razzouk BI: Clinical significance of central nervous system involvement at diagnosis of pediatric acute myeloid leukemia: A single institution's experience. Leukemia. 17:2090–2096. 2003. View Article : Google Scholar : PubMed/NCBI

168 

Lin KI, Tam CS, Keating MJ, Wierda WG, O'Brien S, Lerner S, Coombes KR, Schlette E, Ferrajoli A, Barron LL, et al: Relevance of the immunoglobulin VH somatic mutation status in patients with chronic lymphocytic leukemia treated with fludarabine, cyclophosphamide, and rituximab (FCR) or related chemoimmunotherapy regimens. Blood. 113:3168–3171. 2009. View Article : Google Scholar

169 

Rai KR, Sawitsky A, Cronkite EP, Chanana AD, Levy RN and Pasternack BS: Clinical staging of chronic lymphocytic leukemia. Blood. 46:219–234. 1975. View Article : Google Scholar : PubMed/NCBI

170 

Binet JL, Auquier A, Dighiero G, Chastang C, Piguet H, Goasguen J, Vaugier G, Potron G, Colona P, Oberling F, et al: A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis. Cancer. 48:198–206. 1981. View Article : Google Scholar : PubMed/NCBI

171 

Muñoz L, Lasa A, Carricondo MT, Hernández C, Ubeda J and Nomdedéu JF: Comparative analysis of ZAP-70 expression and Ig VH mutational status in B-cell chronic lymphocytic leukemia. Cytometry B Clin Cytom. 72:96–102. 2006.PubMed/NCBI

172 

Michels E, Vandesompele J, De Preter K, Hoebeeck J, Vermeulen J, Schramm A, Molenaar JJ, Menten B, Marques B, Stallings RL, et al: ArrayCGH-based classification of neuroblastoma into genomic subgroups. Genes Chromosomes Cancer. 46:1098–1108. 2007. View Article : Google Scholar : PubMed/NCBI

173 

Daver N, Schlenk RF, Russell NH and Levis MJ: Targeting FLT3 mutations in AML: Review of current knowledge and evidence. Leukemia. 33:299–312. 2019. View Article : Google Scholar : PubMed/NCBI

174 

Sakaguchi M, Yamaguchi H, Najima Y, Usuki K, Ueki T, Oh I, Mori S, Kawata E, Uoshima N, Kobayashi Y, et al: Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in acute myeloid leukemia. Blood Adv. 2:2744–2754. 2018. View Article : Google Scholar : PubMed/NCBI

175 

Kennedy VE and Smith CC: FLT3 mutations in acute myeloid leukemia: key concepts and emerging controversies. Front Oncol. 10:6128802020. View Article : Google Scholar

176 

Basso G, Veltroni M, Valsecchi MG, Dworzak MN, Ratei R, Silvestri D, Benetello A, Buldini B, Maglia O, Masera G, et al: Risk of relapse of childhood acute lymphoblastic leukemia is predicted by flow cytometric measurement of residual disease on day 15 bone marrow. J Clin Oncol. 27:5168–5174. 2009. View Article : Google Scholar : PubMed/NCBI

177 

Bader P, Kreyenberg H, Henze GH, Eckert C, Reising M, Willasch A, Barth A, Borkhardt A, Peters C, Handgretinger R, et al: Prognostic value of minimal residual disease quantification before allogeneic stem-cell transplantation in relapsed childhood acute lymphoblastic leukemia: The ALL-REZ BFM Study Group. J Clin Oncol. 27:377–384. 2009. View Article : Google Scholar

178 

Jongen-Lavrencic M, Grob T, Hanekamp D, Kavelaars FG, Al Hinai A, Zeilemaker A, Erpelinck-Verschueren CAJ, Gradowska PL, Meijer R, Cloos J, et al: Molecular Minimal Residual Disease in Acute Myeloid Leukemia. N Engl J Med. 378:1189–1199. 2018. View Article : Google Scholar : PubMed/NCBI

179 

Vosberg S and Greif PA: Clonal evolution of acute myeloid leukemia from diagnosis to relapse. Genes Chromosomes Cancer. 58:839–849. 2019. View Article : Google Scholar : PubMed/NCBI

180 

Lussana F, Caprioli C, Stefanoni P, Pavoni C, Spinelli O, Buklijas K, Michelato A, Borleri G, Algarotti A, Micò C, et al: Molecular detection of minimal residual disease before allogeneic stem cell transplantation predicts a high incidence of early relapse in adult patients with NPM1 positive acute myeloid leukemia. Cancers (Basel). 11:14552019. View Article : Google Scholar : PubMed/NCBI

181 

Balatzenko G, Vundinti BR and Margarita G: Correlation between the type of bcr-abl transcripts and blood cell counts in chronic myeloid leukemia-a possible influence of mdr1 gene expression. Hematol Rep. 3:e32011. View Article : Google Scholar

182 

Brinkmann U: Functional Polymorphisms of the Human Multidrug Resistance (MDR1) Gene: Correlation with P glycoprotein Expression and Activity in vivo. Novartis Found Symp. 243:207–10; discussion 207-212. 231-5. 2002. View Article : Google Scholar : PubMed/NCBI

183 

Vendrik CP, Bergers JJ, De Jong WH and Steerenberg PA: Resistance to cytostatic drugs at the cellular level. Cancer Chemother Pharmacol. 29:413–429. 1992. View Article : Google Scholar : PubMed/NCBI

184 

Vandyke K, Fitter S, Dewar AL, Hughes TP and Zannettino ACW: Dysregulation of bone remodeling by imatinib mesylate. Blood. 115:766–774. 2010. View Article : Google Scholar

185 

Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger MW, Silver RT, Goldman JM, Stone RM, et al: Five-Year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 355:2408–2417. 2006. View Article : Google Scholar : PubMed/NCBI

186 

Millot F, Baruchel A, Guilhot J, Petit A, Leblanc T, Bertrand Y, Mazingue F, Lutz P, Vérité C, Berthou C, et al: Imatinib is effective in children with previously untreated chronic myelogenous leukemia in early chronic phase: Results of the French National Phase IV Trial. J Clin Oncol. 29:2827–2832. 2011. View Article : Google Scholar : PubMed/NCBI

187 

Ebnöether M, Stentoft J, Ford J, Buhl L and Gratwohl A: Cerebral oedema as a possible complication of treatment with imatinib. Lancet. 359:1751–1752. 2002. View Article : Google Scholar : PubMed/NCBI

188 

Mughal TI and Schrieber A: Principal long-term adverse effects of imatinib in patients with chronic myeloid leukemia in chronic phase. Biologics. 4:315–323. 2010.

189 

Suttorp M and Millot F: Treatment of pediatric chronic myeloid leukemia in the year 2010: Use of tyrosine kinase inhibitors and stem-cell transplantation. Hematology Am Soc Hematol Educ Program. 2010:368–376. 2010. View Article : Google Scholar

Related Articles

Journal Cover

December-2024
Volume 65 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li J, Wang Y, Dong C and Luo L: Advancements in leukemia management: Bridging diagnosis, prognosis and nanotechnology (Review). Int J Oncol 65: 112, 2024.
APA
Li, J., Wang, Y., Dong, C., & Luo, L. (2024). Advancements in leukemia management: Bridging diagnosis, prognosis and nanotechnology (Review). International Journal of Oncology, 65, 112. https://doi.org/10.3892/ijo.2024.5700
MLA
Li, J., Wang, Y., Dong, C., Luo, L."Advancements in leukemia management: Bridging diagnosis, prognosis and nanotechnology (Review)". International Journal of Oncology 65.6 (2024): 112.
Chicago
Li, J., Wang, Y., Dong, C., Luo, L."Advancements in leukemia management: Bridging diagnosis, prognosis and nanotechnology (Review)". International Journal of Oncology 65, no. 6 (2024): 112. https://doi.org/10.3892/ijo.2024.5700