Published on in Vol 24, No 7 (2022): July

Preprints (earlier versions) of this paper are available at https://preprints.jmir.org/preprint/36490, first published .
A Review of Artificial Intelligence Applications in Hematology Management: Current Practices and Future Prospects

A Review of Artificial Intelligence Applications in Hematology Management: Current Practices and Future Prospects

A Review of Artificial Intelligence Applications in Hematology Management: Current Practices and Future Prospects

Review

1College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar

2National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar

3Surgical Research Section, Department of Surgery, Hamad Medical Corporation, Doha, Qatar

4College of Medicine, Qatar University, Doha, Qatar

5College of Medicine, University of Glasgow, Glasgow, United Kingdom

Corresponding Author:

Adel Elomri, PhD

College of Science and Engineering

Hamad Bin Khalifa University

Education City, Qatar Foundation

Gate 8, Ar-Rayyan

Doha, PO Box 34110

Qatar

Phone: 974 44458536

Email: aelomri@hbku.edu.qa


Background: Machine learning (ML) and deep learning (DL) methods have recently garnered a great deal of attention in the field of cancer research by making a noticeable contribution to the growth of predictive medicine and modern oncological practices. Considerable focus has been particularly directed toward hematologic malignancies because of the complexity in detecting early symptoms. Many patients with blood cancer do not get properly diagnosed until their cancer has reached an advanced stage with limited treatment prospects. Hence, the state-of-the-art revolves around the latest artificial intelligence (AI) applications in hematology management.

Objective: This comprehensive review provides an in-depth analysis of the current AI practices in the field of hematology. Our objective is to explore the ML and DL applications in blood cancer research, with a special focus on the type of hematologic malignancies and the patient’s cancer stage to determine future research directions in blood cancer.

Methods: We searched a set of recognized databases (Scopus, Springer, and Web of Science) using a selected number of keywords. We included studies written in English and published between 2015 and 2021. For each study, we identified the ML and DL techniques used and highlighted the performance of each model.

Results: Using the aforementioned inclusion criteria, the search resulted in 567 papers, of which 144 were selected for review.

Conclusions: The current literature suggests that the application of AI in the field of hematology has generated impressive results in the screening, diagnosis, and treatment stages. Nevertheless, optimizing the patient’s pathway to treatment requires a prior prediction of the malignancy based on the patient’s symptoms or blood records, which is an area that has still not been properly investigated.

J Med Internet Res 2022;24(7):e36490

doi:10.2196/36490

Keywords



Background on Hematologic Malignancies

Blood cancers, namely leukemia and lymphoma, are generally ranked among the most common and deadliest cancer types [1]. Typically, hematologic malignancies result from abnormal growth of white blood cells (WBCs) in the human body, which creates a disproportion among blood elements [2]. The blood contains red blood cells (RBCs), WBCs, and platelets. The role of RBCs is to transmit oxygen from the heart to the entire system, and they constitute the largest proportion of the blood volume [3]. By contrast, WBCs play a crucial role in protecting the body from diseases and infection by deploying various immune mechanisms [4]. Hence, maintaining a healthy WBC level is imperative for the protection of the human body. Normally, these blood elements mature and replenish depending on the body’s needs [5]. However, their growth can become disordered when certain hematologic malignancies are present [6]. For instance, because of the considerable increase in the number of abnormal WBCs, the ability of bone marrow to generate and support healthy RBCs and platelets in terms of oxygen and nutrition supply is impaired [2,7]. Moreover, these malignant WBCs can circulate throughout the body via blood and cause irreparable damage to other organs such as the liver, kidney, and brain [8].

Challenges in Hematology Management

Although complete blood count (CBC) tests often serve as the first step in detecting hematologic malignancies by identifying abnormal blood cell count or any distortion in cell morphology, this simple test is generally deemed insufficient for a practitioner to diagnose blood cancer [5,9]. Therefore, several microscopic evaluations of the blood smear are performed to reach a final diagnosis [5]. As all the available methods are manual and require highly skilled medical personnel for interpretation, a blood cancer diagnosis can be costly and time consuming, which negatively impacts the patient’s efficient and timely treatment [10]. Another challenging aspect in hematology detection is that WBCs are surrounded by other blood components. Thus, the current identification method of manually counting the number of WBCs that appear abnormal does not provide accurate classification results [11]. In fact, it has been reported that diagnostic delays mainly occur because of the complexity of symptom analysis and challenges associated with disease diagnosis.

Artificial Intelligence in Hematology Management

Motivated by the remarkable achievements of artificial intelligence (AI) in various fields, the applicability of such algorithms in solving critical problems related to oncology and hematology was recently investigated and proven efficient [6]. In particular, machine learning (ML) and deep learning (DL) methods were used to assist to classify various cancer types, facilitate faster diagnosis, and provide a basis for accurate clinical decisions for better health outcomes. The 2 main challenges in the implementation of AI in medicine are the limitation and restriction of health information.

This review provides a comprehensive contribution to the hematology care management field, with the objective of studying the applications of AI in various blood cancer stages and detecting the limitations of ML- and DL-based models that have been previously implemented. This paper attempts to highlight the existing gaps in the field of hematology management by studying, classifying, and analyzing 144 papers published between 2015 and 2021.

The “Methods” section clarifies the methodology used to perform the search of all reviewed papers. The “Results” section discusses and classifies existing research based on malignancy type and stage, respectively. Finally, the “Discussion” section provides a detailed analysis of AI applications for hematologic malignancies.


In this review, peer-reviewed and publicly available journal papers were identified from a variety of online databases, while publicly unavailable copies were obtained through our institutional access to journal publications and databases. The search for appropriate journal papers was performed using specific keywords such as AI, ML, blood cancer, hematology, malignancy, leukemia, management, and cancer, which were linked and combined using 2 Boolean operators “AND” and “OR” to produce more focused outcomes (Figure 1).

The paper collection process targeted all the articles written in English in the last 7 years with the aforementioned keywords in their abstracts or titles. The search identified 567 papers, of which only 144 were retained for review. The 423 excluded papers either focused on oncology without paying special attention to hematology, addressed hematologic malignancies from an automated perspective without employing AI models, or covered purely technical drug treatments for blood cancers.

Figure 1. Review chart showing paper elimination and categorization process. AI: artificial intelligence; DL: deep learning; ML: machine learning.
View this figure

Overview

This section presents the paper collection results, as well as an analysis of the literature and its classification with respect to the study category, malignancy type, and pathway stage.

From the analysis of the selected papers, Figure 2 lists the top 10 most globally cited documents in the field of AI in hematology management, with “Intelligent leukemia diagnosis with Bare-bones PSO based feature optimization” [12] rated as the most cited article with 72 citations globally.

Figure 2. Citation statistics for the most cited documents.
View this figure

Descriptive Analysis

With the emergence of AI and the remarkable results it has achieved over the last decade in many areas, researchers in the field have recently started to investigate its applications in blood cancer management. Figure 3 maps the evolution of thematic research in the field of hematology. We can clearly see a spike in the year 2019 in the occurrence of ML and leukemia, followed by DL, AI, and classification trends in the year 2020, which indicates the progression and development of ML and DL themes and their applications in the blood cancer field as well as the recent focus on AI to drive hematologic research.

Furthermore, keywords are used to outline the content of different articles. An analysis of the high-frequency keywords can give an idea about the current research status and potential future directions in the area of AI in hematology management.

Using the mapping and analysis tools provided in the Biblioshiny software (K-Synth Srl), a word tree map was generated to highlight the top 10 most frequently used keywords by scholars in the field. Figure 4 shows the hierarchical visualization of author keywords.

The highest frequencies were for ML (29%), leukemia (15%), classification (12%), and DL (11%), with 32, 17, 13, and 12 occurrences respectively, indicating the recent focus on ML and DL applications in the detection and classification of leukemia and its types (acute myeloid leukemia [AML] and acute lymphoblastic leukemia [ALL]). The tree map also highlights the different techniques and tools used in this field of research through keywords such as “segmentation,” “image processing,” and “flow cytometry,” with an identical frequency of 5% each.

Figure 3. Thematic evolution of hematology management research.
View this figure
Figure 4. Statistics for most cited keywords.
View this figure

Category Selection

In the process of analyzing and classifying the set of collected articles, the generated documents were categorized based on the blood malignancy type, pathway stage, and ML/DL techniques used. The collected papers were either reviews of the literature, journal papers, or conference proceedings. Nonetheless, special attention was paid to review papers, as they provided a clear idea of the research that has been carried out as well as the gaps that remain to be addressed. Table 1 provides a detailed classification of the collected articles by category.

Table 1. Classification of journal papers by category.
CategoriesNumber of articlesStudies
Review papers13[5,13-24]
Conference proceedings33[1,4,6,10,11,25-52]
Journal articles98[2,3,7-9,12,53-144]

Material Evaluation: Hematologic Malignancy Types

After excluding the reviews, this section classifies the 131 remaining journal papers based on the malignancy type and pathway stage.

Although leukemia and lymphoma share some common symptoms, there are major differences in their origins, causes, and treatments. Leukemia is a slowly developing disease that can be either chronic or acute [12]. Acute leukemia spreads quickly, whereas chronic leukemia develops more slowly during its initial stages and is generally more common [33]. For both types, leukemia can be myeloid, which affects the myeloid cells that give rise to WBCs; or lymphoblastic, which starts in cells that later become lymphocytes. Lymphoma, by contrast, affects the lymph nodes. In the case of lymphoma, WBCs (B and T lymphocytes) exhibit abnormal proliferation and based on the presence or absence of Reed-Sternberg cells, they are classified as either Hodgkin or non-Hodgkin diseases, respectively. Table 2 classifies the studies based on the type of malignancies that they address, as either acute myeloid/lymphoblastic leukemia, chronic myeloid/lymphoblastic leukemia, lymphoma, or other less common types of hematologic diseases.

Among the aforementioned malignancy types, AML is the most addressed hematologic disease in the literature with an occurrence of 32.1% (42/131) in the collected journal papers, followed by ALL. Apparently, the applicability of AI methods in managing chronic lymphocytic leukemia (CLL; 13/131, 9.9%) and chronic myeloid leukemia (CML; 2/131, 1.5%) are the least explored areas, wherein further research is pivotal.

Table 2. Distribution of the 131 studies based on malignancy type.
Malignancy typeValues, n (%)
Acute myeloid leukemia42 (32.1)
Acute lymphoblastic leukemia40 (30.5)
Chronic lymphocytic leukemia13 (9.9)
Chronic myeloid leukemia2 (1.5)
Lymphoma17 (13.0)
Other17 (13.0)

Pathway Stages of Hematologic Malignancies

Prediction

The absence of symptoms during the early stages of leukemia makes it challenging for practitioners to predict the occurrence of cancer [117]. Typically, leukemia detection relies on the use of blood cell image classification methods [29]. The introduction of AI-based models is intended to enhance identification accuracy and provide early prediction of potential spread within the human body to help increase the chances of patient recovery and survival.

Screening

No standardized leukemia screening tests have been proven reliable and efficient enough to identify the presence of blood cancer in its early stages [83]. The causes of leukemia remain unknown and a diagnosis cannot be confirmed until the initial symptoms develop. Thus, there is ongoing research on imaging techniques, screening methods, and ways to increase the contribution of AI to provide a better understanding of the root causes of this condition and thus enable expert hematologists to identify and diagnose blood malignancies accurately [56].

Diagnosis

For many diseases, diagnosis is the most crucial stage in the illness pathway [85]. As the field of hematologic research is expanding in an unprecedented manner, the emergence of AI provides an opportunity to overcome the challenge of handling large amounts of imaging data, improve the efficiency and quality of hematologic pattern identification, and provide a clear understanding of suitable therapies following a precise diagnosis [87].

Treatment

AI integration into blood cancer treatment has enabled many advanced practices to deliver timely and efficient therapy [20]. Several techniques were used to identify the doses and combinations of drugs required for each patient as functions of their health status, age, and other essential factors. Table 3 classifies the 131 journal papers based on their corresponding pathway stage.

While there has been more emphasis in the literature on the diagnosis stage, which represents 39.7% (52/131) of the total journal papers collected, treatment and prediction are the least investigated pathway stages with only 13.7% (18/131) and 20.6% (27/131), respectively. This is mainly due to the difficulty in identifying crucial biomarkers that can be used as discriminative feature variables to predict the disease before the onset of symptoms. Similarly, in the case of treatment, as detailed in later sections of this paper, most of the AI-based models [9,42,75] reported in the treatment phase utilize gene expression profiles for model development, which are relatively complex. Hence, more research is needed to investigate the feasibility of easily available biological features for the prediction of treatment-related disease relapse and patient survival.

Table 3. Classification of journal publications by pathway stage (N=131).
Pathway stageValues, n (%)Studies
Prediction27 (20.6) [1,6,7,29,38,40,41,50,51,53,55,64,69,82,92,94,96,99-102,105,117,118,136,143,144]
Screening34 (26.0) [3,10,28,30,32-34,36,44-46,57,58,61,66,71,72,76-78,80,83,85,89,91,106,112,115,119,124,129,130,132,138]
Diagnosis52 (39.7)[2,4,8,11,12,25-27,31,35-37,39,43,47-49,52,54,60,62,63,65,67,68,70,79,84,86-88,90,95,97,98,107,108, 110,111,113,116,120-123,125-128,133,134,137]
Treatment18 (13.7) [9,42,56,59,73-75,81,93,103,104,109,114,131,135,139-141]

This section analyses the existing literature with respect to the methods applied (ML or DL) for solving issues in each pathway stage.

Hematology Prediction

Machine Learning–Based Models

Childhood ALL is a malignant cancer that is the leading cause of pediatric cancer mortality, and around 20% of the children fully treated end up having a recurrence [131]. Hence, it is crucial to predict relapse to deal with the multiple risk groups accordingly. For better management and follow-up planning, Pan et al [131] introduced an ALL relapse prediction model based on ML algorithms that help classify patients with ALL into their appropriate risk categories. In the model selection process, 103 clinical variables were used to train 4 classification algorithms, random forest (RF), decision tree, support vector machine (SVM), and linear regression, to distinguish relapses from nonrelapses in the 3 clinically predefined risk categories: standard-, intermediate-, and high-risk levels. While Pan et al [131] built a model to predict disease relapse, Hauser et al [144] studied the possibility of predicting CML prior to diagnosis using only CBC test results and ML algorithms such as XGBoost and LASSO algorithms on 1623 patients with a definitive CML status. The variables used in the study included laboratory CBC test results, patient demographic features such as their age and gender, and patient encounter information (the number of patient visits to outpatient clinics, etc.). A forward feature selection process was employed to measure the predictive performance of the most potential predictors. The data set was then divided into 7 subsets, in which time of diagnosis was set as a patient baseline, and the 6 remaining sets corresponded to the different time periods preceding the diagnosis test. Interestingly, variable selection yielded different features for inclusion in the models depending on the data collection interval.

The performances of the chosen classifiers in [131] were evaluated by a 10-fold cross validation in each of the 100 training sets. However, the suggested approach is considered insufficient for internal validation that requires at least 50 repeats [145]. By contrast, the chosen data set in [144] was divided into 2 distinct groups: train/validation and test groups. While the latter split-sample validation approach seems reasonable and justifiable to use in this case given the large sample size, potential drawbacks may arise, and several aspects still need attention throughout the application. For instance, as the sample split was performed fully at random, substantial patient imbalances might have occurred with respect to the distributions of predictors and the output. Moreover, 20% was used for model assessment leading to a potential biased evaluation of the model’s performance [145].

Furthermore, it is well known that patients with leukemia often suffer from health issues due to frequent infections, which can lead to death if it is not detected early. Toward this end, Agius et al [55] investigated the risk of infection due to a weakness in the immune system or a cytotoxic treatment immediately after CLL diagnosis by developing the CLL Treatment-Infection Model (CLL-TIM). For each patient, the prediction point was set at 3 months after their diagnosis, and the target output was the 2-year infection risk or CLL treatment. After excluding 74 patients who died and 373 who initiated their treatment before the prediction point, the study cohort’s final size corresponded to 3729 patients. In contrast to Hauser et al [144], Agius et al [55] employed stratified sampling to maintain class distributions and compensate for the 52% International Prognostic Index for CLL (CLL-IPI) missing variables, by dividing the data set into 65% training set and 17.5% for each of the test and internal validation sets. Using 7288 features resulting from a collection of variables from different sources, comprising baseline variables at the time of diagnosis including age, gender, etc.; routine laboratory tests; microbiology findings; pathology reports; and diagnosis codes for all patients, the CLL-TIM ensemble algorithm was composed of 28 ML models that could identify patients at a high risk of infection to increase their chances of survival. The model was then validated on both internal and independent external test cohorts, and exhibited interesting performances, surpassing the CLL-IPI.

Deep Learning–Based Models

Hassouneh et al [50] suggested the use of deep neural networks (DNNs) to predict survivability of patients with leukemia to boost the psychological state of patients and enable physicians to arrange the proper treatments for different cases. The final DNN structure encompassed 6 hidden layers, with 45 hidden neurons in each corresponding hidden layer, and a dropout activation with 25%. While Hassouneh et al [50] used patient records and attributes of patients with leukemia for modeling, Boldú et al [134] relied on a set of 731 blood smear images to predict initial patient diagnosis. For data set limitation purposes, 4 convolutional neural network (CNN) models were pretrained on a larger data set and used for the aforementioned task. Their respective performances were compared and the architecture achieving the best outcome was trained further. Next, these pretrained CNNs were fine-tuned to match the type of data that are fed into the model. Finally, the proposed ALNet model was able to distinguish, on a first level, between healthy and abnormal blood cell images. On a second level, it was able to identify whether the blasts were myeloid or lymphoid. After a thorough evaluation process using 5-fold cross validation and the hold-out (80%/20%) approach with 470 iterations, ALNet demonstrated interesting results and was chosen as the best architecture for modeling. One strength of this study is that the 5-fold cross validations were approximately balanced, and despite the random split, the data from the same patient smear were maintained within the same fold. Alternatively, the proposed DNN algorithm in [50] was trained and evaluated using 2 methods: 10-fold cross validation and ensemble method. Although the 10-fold cross-validation method is known to generate more stable results, the number of repetitions is quite crucial, which was absent in this study [145].

Overall, it is very clear that all the aforementioned algorithms succeeded in predicting the different aspects and repercussions of the disease. Nevertheless, no existing literature has yet examined the root cause of the malignancy by predicting the possibility of patient infection. Table 4 summarizes some studies in the prediction phase alongside their objectives, the data sets used in the studies, the methodologies followed, the performance of the models applied, their strengths and weaknesses, and the validation approach used, where available.

Table 4. Study analysis for journal publications on the prediction phase.
ReferenceObjective, data set, and methodologyPerformance and remarks
[131]
  • Objective: ALLa relapse prediction
  • Data set: 336 newly diagnosed children with ALL
  • Methodology: Random forest algorithm
Performance:
  • Accuracy: 0.829
  • AUCb: 0.902
Strengths:
  • Usage of 4 MLc algorithms and 104 features
  • Good model performance in all risk-level groups
  • Adoption of a special feature selection strategy: 100-fold Monte Carlo cross validation combined with 10-fold cross validation
Limitations:
  • Data set imbalance (relapsed and nonrelapsed children)
  • Strong predictors were excluded from the variable set
Validation:
  • 10-fold cross validation
[144]
  • Objective: Prediction of patients with CMLd and non-CML using complete blood count records
  • Data set: Complete blood count records of 1623 patients with a BCR-ABL1 test extracted from the US Veterans Health Administration
  • Methodology: XGBoost and LASSO
Performance:
  • AUC range: 0.87-0.96 at the time of diagnosis
Strengths:
  • Use of 2 models
  • Use of 2 feature selection methods
Limitations:
  • Imbalanced data set (predominant gender is male)
  • Nonstandard data collection process
Validation:
  • Split sample validation (20% of the data for validation)
[1]
  • Objective: Leukemia detection based on biomedical data
  • Data set: 401 leukemia datapoints from Z H Sikder Medical College and Hospital
  • Methodology: Decision tree
Performance:
  • Accuracy: 100%
Strengths:
  • Use of 4 supervised ML algorithms
Limitations:
  • Overfitting
Validation:
  • 10-fold cross validation
[50]
  • Objective: Prediction of leukemia survivability
  • Data set: 131,615 records and 133 attributes for patients with leukemia from the SEERe database
  • Methodology: Deep neural network model
Performance:
  • Accuracy: 74.85%
Strengths:
  • Use of a DNNf ensemble method
Limitations:
  • Many problems in the leukemia data set (redundant attributes, missing values, and unknown values)
Validation:
  • 10-fold cross validation
  • Ensemble method
[96]
  • Objective: Predictive identification of patients at risk during treatment
  • Data set: 737 samples of patients diagnosed with CLLg at Mayo Clinic
  • Methodology: logistic regression, support vector machine, gradient boosting machine, random forest
Performance:
  • ROCh-AUC: above 80%
Strengths:
  • Binary classification outperforms survival analytic methods
Limitations:
  • Lack of actionable information provided by the ML algorithms
Validation:
  • 100 runs of 5-fold cross validation

aALL: acute lymphoblastic leukemia.

bAUC: area under the curve.

cML: machine learning.

dCML: chronic myeloid leukemia.

eSEER: Surveillance, Epidemiology, and End Results

fDNN: deep neural network.

gCLL: chronic lymphocytic leukemia.

hROC: receiver operating characteristic

Hematology Screening

Machine Learning–Based Models

Initial leukemia screening and its efficient diagnosis require a deep and thorough image analysis process. As opposed to traditional manual screening, automated leukemia screening is a novel approach that minimizes human interaction and provides more accurate clinical information by using blood smear images to identify ALL automatically [17]. The automated screening process is considered challenging due to the leukocyte localization and region extraction phases, which are generally obtained via background removal and separation of surrounding blood components that might distort the overall detection process. For this reason, many studies have employed techniques such as principal component analysis as a filter to identify any features that do not bring any important information to the classification process [115] to enhance detection accuracy. Similarly, Chebouba et al [32] used a meta-heuristic stochastic local search technique to select the most important genes and proteins to be used in the RF-based classification of patients with AML.

Deep Learning–Based Models

CML consists of 3 sequential phases that change based on the patient’s status and can progress to more severe phases if timely treatment is not provided. This makes CML phase identification very crucial, as different phases require separate treatments and medical regimens. In the chronic phase, less than 10% of the cells in both the blood and bone marrow are blasted. The severity and persistence of the aforementioned phase depend mainly on the consistency of the therapy followed. If the chronic stage is neglected and the patient does not receive timely and effective treatment, the condition can deteriorate to reach an accelerated phase where the blast count increases to around 10%-19%. Similarly, if the patient’s condition declines with no appropriate medical intervention, the percentage of blasted WBCs doubles to reach around 20% or more. At this stage, the patient’s state is considered uncontrollable, and the patient starts to exhibit symptoms such as fever, weakness, and weight loss [33]. As CNNs were proven to be efficient tools for accurate image recognition, Khosla and Ramesh [33] suggested using the latter to classify different CML images into their respective phases. Similarly, Togacar et al [71] used CNNs to separate WBC images into their 4 subclasses: eosinophil, lymphocyte, monocyte, and neutrophil. While the aforementioned CNNs are successful in identifying the most important features in images with no human supervision, they generally require large training data to achieve high performance, and their employment is regarded expensive in terms of both time and training. For this reason, while many studies tend to employ image augmentation to the existing data set to create larger samples by slightly changing the existing collected images [33], others implement the concept of transfer learning to overcome the training data shortage. The idea behind the latter is to leverage the power and knowledge of a pretrained model to apply it on a new similar task. For instance, Sahlol et al [3] proposed a novel approach consisting of a hybrid model and combined CNN feature extraction using the solid architecture of VGGNet that was pretrained on ImageNet, to separate malignant cells from benign ones. Similarly, Li et al [127] developed the globally optimized transfer deep-learning platform with multiple pretrained CNNs (GOTDP-MP-CNNs). This DL platform is composed of 17 CNNs able to classify pathologic images into human diffuse large B-cell lymphoma and non–diffuse large B-cell lymphoma. Table 5 summarizes some of the collected studies in the screening phase.

Table 5. Study analysis for journal papers in the screening phase.
ReferenceObjective, data set, and methodologyPerformance and remarks
[3]
  • Objective: Classification of white blood cell leukemia
  • Data set: Acute Lymphoblastic Leukemia Image Database for Image Processing 1 and 2
  • Methodology: A hybrid model (CNNa and SESSAb)
Performance:
  • Accuracy: 99.2%
  • Sensitivity: 100%
Strengths:
  • Powerful performance using CNN
  • Use of the salp swarm optimization method
  • Hybrid classification method
  • Use of transfer learning
Limitations:
  • Small limited data set insufficient to train CNNs
Validation:
  • 5-fold internal cross validation and 20% testing (external validation)
[61]
  • Objective: Automated identification of acute lymphoblastic leukemia
  • Data set: Blood smear images obtained from the Department of Hematology at the University Hospital Ostrava
  • Methodology: support vector machine/artificial neural networks
Performance:
  • Accuracy: 98.19%
Strengths:
  • High classification accuracy
  • Successful feature selection
Limitations:
  • Extensive preprocessing is required
  • Lack of medical data sets
  • Inability to generalize the results and trends for lack of comparison with other methods
Validation:
  • 10-fold cross validation repeated 10 times
[33]
  • Objective: Classification of chronic myeloid leukemia phases
  • Data set: 500 pictures from Patliputra Medical College and Hospital, Dhanbad, and the blood journal repository
  • Methodology: CNN
Performance:
  • Accuracy: 97.8%
Strengths:
  • Use of transfer learning
Limitations:
  • Limited data set
Validation:
  • Internal validation (14 left for testing)

aCNN: convolutional neural network.

bSESSA: statistically enhanced salp swarm algorithm.

Hematology Diagnosis

Machine Learning–Based Models

To address the challenge of manually detecting blasted cells, Dasariraju et al [54], Inbarani et al [66], Abedy et al [29], Jagadev and Virani [34], and Dharani and Hariprasath [31] used medical images of healthy and malignant samples to automatically identify the leukemic types and subtypes. While Dasariraju et al [54] applied an RF algorithm as an approach to differentiate between abnormal and healthy leukocytes, and classify immature leukocytes into their 4 subtypes, Inbarani et al [66] discussed the implementation of a novel sophisticated approach to identify ALL blast cells via the histogram-based soft covering rough K-means clustering (HSCRKM) segmentation algorithm. The latter is a hybrid-clustering technique that combines the strengths of both the soft covering rough set and the rough K-means clustering. Nevertheless, one main limitation of the HSCRKM segmentation technique is that it is not suitable for multiple color images because the latter increase the processing time due to an increase in the peak values of the histogram. To enhance image representation and prepare a clean input for the classification model, Jagadev and Virani [34] applied SVM on 220 blood smear images of healthy individuals and patients with leukemia to identify the 4 leukemic subtypes (AML, CML, CLL, and ALL) using both K-means clustering and hue, saturation, value color–based segmentation techniques. Similarly, Dasariraju et al [54] performed a set of morphological imaging modifications and preprocessing techniques to segment the nucleus and cytoplasm and overcome the difficulty of blood image detection. To reduce data dimensionality, the model’s speed, and processing time, the most relevant features were obtained using feature extraction techniques and the resulting output was passed on to the classifier [34,54,66]. Alternatively, Abedy et al [29] chose to employ the histogram of oriented gradients for feature extraction, the Gaussian filter for noise elimination, and the Sobel kernel for image filtering. Furthermore, Dey and Islam [49] adopted the principal component analysis technique followed by grid search for hyperparameter tuning, which significantly decreased the number of components from 7129 features to only 6 important parameters. This data transformation technique did not only reduce the computational time and made it faster, but it also helped giving better results. Alongside the usage of imaging techniques, many studies employed other techniques; for example, Moraes et al [133] suggested the usage of flow cytometry data for distinguishing leukemia/lymphoma, and Mahmood et al [143] directed their research to focus more on identifying the most discriminatory features for CLL using patient laboratory test results, demographic parameters, and training a Classification and Regression Trees model on 94 pediatric patients, which was evaluated using 10-fold cross validation. Moreover, both Dharani and Hariprasath [31] and Jagadev and Virani [34] used SVM to classify leukemia and its subtypes, while Paswan and Rathore [28] used K-nearest neighbors to separate blasted blood cells from normal ones and classify them further into either AML or ALL using a value of K=4. By contrast, Moraes et al [133] suggested the implementation of decision tree as an ML-based technique for distinguishing leukemia/lymphoma, where a binary classification between healthy and immature leukocytes was performed with an 80%/20% data split, followed by a subclassification of immature leukocytes into their respective 4 types using a 70%/30% split, and several combinations of hyperparameters were evaluated during a 5-fold cross validation. Conversely, Abedy et al [29] chose to employ logistic regression as a classifier to identify the shape of the leukemic cell from microscopic blood images, while Dey and Islam [49] conducted a study to detect patients’ leukemia type based on their gene expression information using the RF algorithm and 2 other algorithms, XGBoost and artificial neural networks (ANNs), and Dasariraju et al [54] used RF to perform a subclassification of immature leukocytes into their respective 4 types.

Deep Learning–Based Models

With the objective of optimizing leukemia diagnosis, some studies made use of genetic features. For example, Rodrigues and Deusdado [63] suggested the application of a kernel logistic regression that classifies gene expression data using meta-learners to select the most relevant attributes before classification. The data set used for training comprised the 2 types of leukemia (ALL and AML) and a set of gene features. Pearson correlation and chi-square statistic were the 2 approaches used on meta-learners to assess attributes. Then, all models used 10-fold cross validation resulting in an identification of 12 common genes. Other studies, such as that by Al-Dulaimi et al [23], focused on current practices, techniques, and challenges in digital hematology detection of WBCs and their components (nuclei and cytoplasm) using hematologic microscopy images. In addition to analyzing the growing trends in computer-aided diagnosis applications, the review highlighted the main challenges associated with the use of CNNs in terms of both high computational time and costs to classify images and detect abnormalities. This gave rise to the use of transfer learning and enhancing optimization techniques, such as the bare bones particle swarm optimization algorithm used by Srisukkham et al [12] to extract the most informative features and enhance the classification accuracy of the lymphocytic cells into either normal or blasted. Likewise, Miyoshi et al [122] directed their work toward enhancing lymphoma diagnosis by classifying histopathological lymphoma images using a DL model. The aim of the study was to evaluate the performance of the suggested automated DL model and compare it with that of a traditional manual hematopathology detection procedure using CNNs. In this study, each test set comprised a total of 100 image patches, and the rest was randomly divided into 5 separate groups. During each repetition for 5 iterations, 1 group was kept for validation to evaluate the classifier performance for every epoch, while the other 4 groups were used for training. The number of epochs used in this study was 30. Similarly, Shafique and Tehsin [125] suggested the deployment of deep CNNs to detect the ALL type and its corresponding subtypes, while Zhao et al [60] proposed turning the captured raw multiparameter flow cytometry data into a 2D image by means of a self-organizing map (SOM) to analyze and classify them using the aforementioned algorithms. An SOM is a map of neurons that relies on unsupervised learning, in the sense that human intervention is not necessarily required. This model is used in many applications and has strong generalization abilities. The model was trained for 15 epochs, at a learning rate of 0.001 using an Adam optimizer function. The evaluation of classification accuracies used a 10% validation split, which was also employed for network architecture optimization. Then, the model performance was evaluated using the hold-out test set. In the same context, Sipes and Li [4] attempted fine-grained image classification for ALL diagnosis and compared the accuracies of CNNs and other models that used specific hand-selected features, while Vincent et al [37] proposed a leukemia classification that could be performed on 2 levels. The first process was applied to well-segmented nuclei extracted from 100 blood smear images. In this step, 90 samples were used for training and 10 were kept for validation. The cells were classified into normal and abnormal. The second step consisted of feeding 5 features extracted from abnormal images into a second classifier that split the images into ALL and AML types accordingly. Table 6 summarizes some of the studies in the diagnosis phase based on their objectives, data sets used, methodologies, and performance characteristics.

Table 6. Study analysis for journal publications on the diagnosis phase.
ReferenceObjective, data set, and methodologyPerformance and remarks
[60]
  • Objective: Classification of mature B-cell neoplasm
  • Data set: 20,622 routine diagnostic samples from Munich Leukemia Laboratory
  • Methodology: CNN-SOMa transformation
Performance:
  • Accuracy: 95%
Strengths:
  • Large data set
  • High accuracy
Limitations:
  • Nonuniform distribution of misclassifications due to similarity in flow cytometric profiles
Validation:
  • 10% validation split
[54]
  • Objective: Detection of immature leukocytes and their classification into 4 types
  • Data set: Images extracted from a publicly available data set at The Cancer Imaging Archive
  • Methodology: Random forest algorithm
Performance:
  • Accuracy: 92.99%
Strengths:
  • High precision results for each class
Limitations:
  • High number of false positives leading to low precision and specificity
Validation:
  • 5-fold cross validation
[49]
  • Objective: Identification of the leukemia type based on patient genetic expression
  • Data set: A sample of 7129 genes that represent the genetic expressions of 72 people from Kaggle
  • Methodology: XGBoost, artificial neural networks, and random forest algorithm
Performance:
  • Random forest accuracy: 80.8%
  • XGBoost accuracy: 92.3%
Strengths:
  • Use of principal component analysis for dimensionality reduction and faster computation
  • Use of grid search for the best hyperparameter selection
Limitations:
  • Small data set (72 people)
Validation:
  • Internal validation (65%/35% split)
[135]
  • Objective: Classification of lymphocytic cells
  • Data set: The ALL-IDB2 Database
  • Methodology: bare bones particle swarm optimization–based feature optimization
Performance:
  • Accuracy: 94.94%-96.25%
Strengths:
  • A good performance on capturing prognostic chronic myeloid leukemia markers by the model
Limitations:
  • Challenge of capturing relationships between data types with no information loss in clinical clustering
Validation:
  • Validation on an external independent clinical trial
[34]
  • Objective: Detection of leukemia and its types
  • Data set: 220 blood smear images from healthy individuals and patients with leukemia
  • Methodology: support vector machine
Performance:
  • Accuracy: Above 80%
Strengths:
  • Use of 3 segmentation methods
  • Broader range of leukemia classification (types and subtypes)
Limitations:
  • Costly method based on imaging data
Validation:
  • Internal validation (train test split)
[122]
  • Objective: Automated detection of malignant lymphoma
  • Data set: Prepared histopathologic images (388 sections, 259 diffuse large B-cell lymphomas, 89 follicular lymphomas, and 40 reactive lymphoid hyperplasia)
  • Methodology: Deep neural network classifier
Performance:
  • Accuracy: 97%
Strengths:
  • High accuracy outperforming 7 pathologists
  • Model ensemble comprising 3 classifiers
Limitations:
  • Classifier requires a manual annotation
  • Model not able to classify all the subtypes
Validation:
  • K-fold cross validation repeated 5 times
[37]
  • Objective: Multiclassification of leukemia
  • Data set: 100 blood smear images
  • Methodology: Neural network classifiers
Performance:
  • Accuracy: 97.7%
Strengths:
  • Two-step neural network classifier
Limitations:
  • Limited data set (100 blood smear images)
Validation:
  • Internal validation (90 images used for training and 10 kept for validation)
[133]
  • Objective: Leukemia and lymphoma diagnosis
  • Data set: 283 blood and bone marrow sample images from patients with leukemia and lymphoma
  • Methodology: Decision tree
Performance:
  • Correctness: 95%
Strengths:
  • Application of the LASSO algorithm for regularization
  • Model robustness and strength against false negatives
Limitations:
  • Complexity of the decision tree and the risk of overfitting through the production of too large trees
Validation:
  • 30-fold cross validation
[66]
  • Objective: Leukemia image segmentation
  • Data set: The Acute Lymphoblastic Leukemia Image Database
  • Methodology: HSCRKMb/particle swarm optimization/K-means
Performance:
  • Accuracy: 80% and above
Strengths:
  • Use of 7 machine learning methods
  • Application of soft covering rough approximation
Limitations:
  • Suitable for medical images only
  • Application on multiple color images increases the processing time
Validation:
  • Different train/test sizes were used for model evaluation
[144]
  • Objective: Determining the most predictive features for acute lymphoblastic leukemia identification
  • Data set: 94 pediatric patient samples collected from the Department of Hematology and Oncology, Children Hospital and Institute of Child Health, Lahore
  • Methodology: Random forest, boosting machine, C5.0 decision tree, and classification and regression trees
Performance:
  • Accuracy: 87.4%
Strengths:
  • High accuracy
  • Balanced data set
Limitations:
  • Small-scale study
  • Few machine learning models
  • Socioeconomic risk factors not selected automatically
Validation:
  • Internal validation (train/validation data)
  • 10-fold cross validation
[31]
  • Objective: Leukemia diagnosis and its subtypes
  • Data set: 200 blood smear images extracted from Vidyalankar Institute of Technology, Mumbai and online databases
  • Methodology: support vector machine
Performance:
  • Accuracy: 97.8%
Strengths:
  • Good detection accuracy
  • Thorough image segmentation process
Limitations:
  • Challenging detection process due to the irregularity of the cancer cell’s shape and nucleus
  • Use of only support vector machine for classification

aCNN-SOM: convolutional neural network-self-organizing map.

bHSCRKM: histogram-based soft covering rough K-means clustering.

Hematology Treatment

Machine Learning–Based Models

Complete remission (CR) refers to the disappearance of all signs and symptoms of an illness [51]. However, a significant proportion of patients report disease relapse after therapy and complete disease recovery. In this regard, Gal et al [75] proposed an ML technique that uses gene expressions to predict the likelihood of CR in patients with AML who previously received therapy. The 473 collected samples were divided into training and testing and were fed into the 3 classifiers for feature selection using a 5-fold cross validation. To select the most significant genes that clearly mark the difference between the state of CR and non-CR, a statistical t test was performed in each fold for each method. For further gene feature selection and performance enhancement, the results were compared with those of 3 algorithms: randomized LASSO, recursive feature elimination, and hill climbing. It was proven that cancers that appear pathologically identical do not necessarily exhibit the exact response to similar drugs. To overcome the challenge of high demands for personalized or patient-specific medicine, Lee et al [9] proposed using a gene-expression profile and in vitro drug sensitivity data to spot molecular markers that explain this patient-specific drug response. The data set used comprised 160 chemotherapy drugs and inhibitors for 30 patients with AML. The gene-drug association was identified using the MERGE algorithm, which utilized gene characteristics such as a novel mutation, expression hubness, known regulator, genomic copy number variation, and methylation. The model testing was done in 2 different ways: the first approach used 2 batches containing 12 patient samples for training and 12 different samples for validation, respectively. The second approach used a leave-one-out cross validation to test the predicted drug sensitivity for 30 patient samples. The latter cross-validation method is regarded as reasonable because it prevents the high computational training cost and time owing to the small sample size and is much less biased than using a single test set because the process fits the data set consisting of n-1 observations repeatedly [145]. Although having a limited data size permitted the obtention of efficient validation results, identifying gene-drug associations is deemed to be a challenging process in that case. However, the proposed algorithm was successful to prioritize genes based on the multidimensional data on their potential to drive cancer. Consequently, upon comparison with other alternate methods (ElasticNet, multitask learning, Pearson P value, and Spearman P value), MERGE exhibited the best gene-drug association result.

Deep Learning–Based Models

In their review and analysis of current AI applications for hematologic disorders’ treatment, Muhsen et al [24] presented a study that was performed with a set of patients who underwent allogeneic hematopoietic cell transplantation to predict the development of acute graft-versus-host disease using ANNs. After comparing the performance acquired by ANNs and the results achieved by logistic regression, ANNs were found to predict the presence of graft-versus-host disease significantly better. However, among the limitations remaining to help reach optimal ML results is the limited data input from patients that could be further enlarged to include biologic and genetic factors, for instance. Moreover, the employment of several sampling techniques such as random oversampling, synthetic oversampling, and remote under sampling could help improve the ML models’ accuracies in predicting treatment-related mortality in allogeneic hematopoietic cell transplantation.

Similarly, Lyu et al [42] used ANNs to classify the progress and change in gene expressions and peripheral blood mononuclear cells before treatment and after starting therapy. Table 7 shows some extracted literature focusing on the hematology treatment phase.

Table 7. Study analysis for journal publications on the treatment phase.
ReferenceObjective, data set, and methodologyPerformance and remarks
[9]
  • Objective: Digital analysis of blood smears and preclassification of cells
  • Data set: Images of blood smears from a hematologic laboratory
  • Methodology: MERGE algorithm
Performance:
  • Accuracy: 90%
Strengths:
  • Introduction of a new computational and statistical method to determine gene markers
Limitations:
  • Small data set comprising only 30 patients with acute myeloid leukemia
Validation:
  • Leave-one-out cross validation
[75]
  • Objective: Prediction of complete remission of acute myeloid leukemia
  • Data set: 473 bone marrow samples from the Children’s Oncology Group
  • Methodology: K-nearest neighbor, support vector machine, and hill climbing
Performance:
  • Area under the curve: 0.84
Strengths:
  • Use of 3 feature selection algorithms: randomized LASSO, recursive feature elimination, and hill climbing
  • Use of 3 classifiers: support vector machine, random forest, and K-nearest neighbor
Limitations:
  • Small data set
Validation:
  • 100 iterations of a 5-fold cross validation
[81]
  • Objective: Identify the right patterns to improve risk stratification of patient with CLLsa
  • Data set: (1) the first cohort comprised CLL cells of 196 individuals; the second cohort comprised CLL cells of 98 individuals including their clinical data and RNA-seq
  • Methodology: (1) EM algorithm and the Gaussian mixture models; (2) Boosted tree ensemble method
Performance:
  • Precision: 90%
Strengths:
  • High accuracy and precision
Limitations:
  • Large data set and 5-year monitoring is required
Validation:
  • External validation on an independent cohort

aCLL: chronic lymphocytic leukemia.

Conclusions and Future Research Directions

Early diagnosis and prediction of hematologic malignancies can immensely reduce mortality rates and can improve patient survival rates. Nevertheless, the nature of data on medical treatment is complex and requires an in-depth analysis to extract the important explicative features and hidden data patterns. The only way to handle enormous sets is through the use of AI. The challenge that faces AI applications, however, is the limitation in data availability, which can be overcome by means of data augmentation techniques, regularization, and transfer learning. This review of the literature highlights the most recent applications of both DL and ML in the field of blood cancer management for every hematologic pathway stage and malignancy type. Based on the reviewed articles, ML techniques have been widely used, in comparison with DL methods, as the latter are relatively newer and require larger data sets than ML, which is considered a constraint in the medical field. In some studies, ML techniques performed better than DL methods and vice versa, depending on the application and nature of data used. Moreover, screening and diagnosis are challenging tasks, as hematologic cancers are difficult to identify during their initial stages. Therefore, many studies in the field investigated the aforementioned stages alongside prediction, while less attention has been paid to the treatment stage. The latter is critical and requires further analysis and study, as repercussions and relapses may arise due to cancer treatment, namely, chemotherapy, which requires a risk evaluation and future mitigation plans. Furthermore, some malignancies appeared to be more addressed than others, mainly acute myeloid/lymphoblastic leukemias that have gotten the most attention in the last few years followed by lymphoma, due to their fast development and aggressivity. Conversely, there was less emphasis and minor existing literature tackling the chronic types of leukemia. This can be due to the slow-growing pattern of the aforementioned, and a lack of sudden symptom exhibition until very late stages, which makes the latter’s monitoring quite complex. Overall, a lack of detection accuracy can have a significant impact on the patient’s journey to treatment because it can delay diagnosis and affect the efficiency of therapies. Therefore, predictive models that can recognize disease patterns and common symptoms in hematologic malignancies based on medical patient records are essential to forecast the risk of infection and avoid late-stage diagnoses. Thus far, many studies employed several techniques to predict hematologic malignancies’ diagnosis through either medical image recognition, flow cytometry, or genetic expressions. However, no study in the literature has ever made use of patient CBC test results alone for blood disorder prediction or detection purposes. As the latter is generally regarded as the first diagnostic routine for hematologists to confirm leukemia diagnosis, it can be an efficient medium to potentially investigate in the future.

Acknowledgments

This article was made possible by the National Priorities Research Program-Standard (NPRP-S) Twelfth (12th) Cycle grant (NPRP12S-0219-190108) from the Qatar National Research Fund (a member of Qatar Foundation). The findings herein reflect the work, and are solely the responsibility, of the author[s].

Conflicts of Interest

None declared.

  1. Das M, Kumar Saha P. A Comprehensive Study of Machine Learning algorithms for Predicting Leukemia Based on Biomedical Data. 2020 Dec 21 Presented at: International Conference on Innovation in Engineering and Technology; December 23-24, 2019; Dhaka, Bangladesh p. 1-5. [CrossRef]
  2. Acharya V, Ravi V, Pham TD, Chakraborty C. Peripheral Blood Smear Analysis Using Automated Computer-Aided Diagnosis System to Identify Acute Myeloid Leukemia. IEEE Trans. Eng. Manage 2021:1-14. [CrossRef]
  3. Sahlol AT, Kollmannsberger P, Ewees AA. Efficient Classification of White Blood Cell Leukemia with Improved Swarm Optimization of Deep Features. Sci Rep 2020 Feb 13;10(1):2536 [FREE Full text] [CrossRef] [Medline]
  4. Sipes R, Li D. Using Convolutional Neural Networks for Automated Fine Grained Image Classification of Acute Lymphoblastic Leukemia. 2019 May Presented at: 2018 3rd International Conference on Computational Intelligence and Applications (ICCIA); July 28-30, 2018; Hong Kong, China p. 157-161. [CrossRef]
  5. Ratley A, Minj J, Patre P. Leukemia Disease Detection and Classification Using Machine Learning Approaches: A Review. 2020 Presented at: 2020 First International Conference on Power, Control and Computing Technologies (ICPC2T); January 3-5, 2020; Raipur, Chhattisgarh, India p. 161-165. [CrossRef]
  6. Schenone D, Dominietto A, Campi C, Frassoni F, Cea M, Aquino S, et al. Radiomics and Artificial Intelligence for Outcome Prediction in Multiple Myeloma Patients Undergoing Autologous Transplantation: A Feasibility Study with CT Data. Diagnostics (Basel) 2021 Sep 24;11(10):1759 [FREE Full text] [CrossRef] [Medline]
  7. Shouval R, Labopin M, Bondi O, Shamay H, Shimoni A, Ciceri F. Prediction of allogeneic hematopoietic stem cell transplantation (Allo-HSCT) related mortality in acute leukemia: Generation of a machine learning-based model using the data set of the Acute Leukemia Working Party (ALWP) of the EBMT. Blood 2013;122(21):409. [CrossRef]
  8. Renuka T, Surekha B. Acute-lymphoblastic leukemia detection through deep transfer learning approach of Neural Network. 2021 Mar 19 Presented at: First Doctoral Symposium on Natural Computing Research; August 8, 2020; Pune, Maharashtra, India p. 163-170. [CrossRef]
  9. Lee S, Celik S, Logsdon BA, Lundberg SM, Martins TJ, Oehler VG, et al. A machine learning approach to integrate big data for precision medicine in acute myeloid leukemia. Nat Commun 2018 Jan 03;9(1):42 [FREE Full text] [CrossRef] [Medline]
  10. Avalos M, Touchais H, Henríquez-Henríquez M. Optimising criteria for Manual Smear Review following Automated Blood Count Analysis: A machine learning approach. Advances in Intelligent Systems and Computing 2021 Apr 10;91:380-391. [CrossRef]
  11. Darane B, Rajput P, Sondagar Y, Koshy R. Recognizing Presence of Hematological Disease using Deep Learning. 2020 Mar 12 Presented at: 2019 Third International conference on I-SMAC (IoT in Social, Mobile, Analytics and Cloud) (I-SMAC); December 12-14, 2019; Palladam, Tamil Nadu, India p. 310-315. [CrossRef]
  12. Srisukkham W, Zhang L, Neoh SC, Todryk S, Lim CP. Intelligent leukaemia diagnosis with bare-bones PSO based feature optimization. Applied Soft Computing 2017 Jul;56:405-419. [CrossRef]
  13. Radakovich N, Cortese M, Nazha A. Acute myeloid leukemia and artificial intelligence, algorithms and new scores. Best Pract Res Clin Haematol 2020 Sep;33(3):101192 [FREE Full text] [CrossRef] [Medline]
  14. Rodellar J, Alférez S, Acevedo A, Molina A, Merino A. Image processing and machine learning in the morphological analysis of blood cells. Int J Lab Hem 2018 May 09;40:46-53. [CrossRef]
  15. Salah HT, Muhsen IN, Salama ME, Owaidah T, Hashmi SK. Machine learning applications in the diagnosis of leukemia: Current trends and future directions. Int J Lab Hematol 2019 Dec 09;41(6):717-725. [CrossRef] [Medline]
  16. Kratz A, Lee S, Zini G, Riedl JA, Hur M, Machin S, International Council for Standardization in Haematology. Digital morphology analyzers in hematology: ICSH review and recommendations. Int J Lab Hematol 2019 Aug 02;41(4):437-447. [CrossRef] [Medline]
  17. Benard B, Gentles AJ, Köhnke T, Majeti R, Thomas D. Data mining for mutation-specific targets in acute myeloid leukemia. Leukemia 2019 Apr 6;33(4):826-843 [FREE Full text] [CrossRef] [Medline]
  18. Banjar H, Adelson D, Brown F, Chaudhri N. Intelligent Techniques Using Molecular Data Analysis in Leukaemia: An Opportunity for Personalized Medicine Support System. BioMed Research International 2017 Jul 25;2017:1-21. [CrossRef]
  19. El Achi HE, Khoury JD. Artificial Intelligence and Digital Microscopy Applications in Diagnostic Hematopathology. Cancers (Basel) 2020 Mar 26;12(4):797 [FREE Full text] [CrossRef] [Medline]
  20. Eckardt J, Bornhäuser M, Wendt K, Middeke J. Application of machine learning in the management of acute myeloid leukemia: current practice and future prospects. Blood Adv 2020 Dec 08;4(23):6077-6085 [FREE Full text] [CrossRef] [Medline]
  21. Umamaheswari D, Geetha S. Review on image segmentation techniques incorporated with machine learning in the scrutinization of leukemic Microscopic Stained Blood Smear Images. 2019 Jan 02 Presented at: International Conference on ISMAC in Computational Vision and Bio-Engineering (ISMAC-CVB); May 2018; Palladam, Tamil Nadu, India p. 1773-1791. [CrossRef]
  22. Khan S, Sajjad M, Hussain T, Ullah A, Imran AS. A Review on Traditional Machine Learning and Deep Learning Models for WBCs Classification in Blood Smear Images. IEEE Access 2021;9:10657-10673. [CrossRef]
  23. AL-Dulaimi K, Banks J, Nugyen K, Al-Sabaawi A, Tomeo-Reyes I, Chandran V. Segmentation of White Blood Cell, Nucleus and Cytoplasm in Digital Haematology Microscope Images: A Review–Challenges, Current and Future Potential Techniques. IEEE Rev. Biomed. Eng 2021;14:290-306. [CrossRef]
  24. Muhsen IN, ElHassan T, Hashmi SK. Artificial Intelligence Approaches in Hematopoietic Cell Transplantation: A Review of the Current Status and Future Directions. Turk J Haematol 2018 Aug 03;35(3):152-157 [FREE Full text] [CrossRef] [Medline]
  25. Alagu S, Bagan K. Computer Assisted Classification Framework for detection of acute myeloid leukemia in Peripheral Blood Smear Images. In: Innovations in Computational Intelligence and Computer Vision. Singapore City, Singapore: Springer; Sep 22, 2020:403-410.
  26. Akter HM, Islam SM, Muntasir I, Muzahidul IA, Islam S, Ahmed A. Leukemia detection mechanism through microscopic image and ML Techniques. 2020 Dec 22 Presented at: 2020 IEEE Region 10 Conference (TENCON); November 16-19, 2020; Osaka, Japan. [CrossRef]
  27. Mandal S, Daivajna V, Rajagopalan V. Machine Learning based System for Automatic Detection of Leukemia Cancer Cell. 2020 Mar 12 Presented at: 2019 IEEE 16th India Council International Conference (INDICON); December 13-15, 2019; Rajkot, Gujarat, India p. 1-4. [CrossRef]
  28. Paswan S, Rathore Y. Recognition and Arrangement of Blood Cancer from Microscopic Cell Pictures Utilizing Support Vector Machine K-Nearest Neighbor and Deep Learning. 2019 Mar 18 Presented at: 2018 International Conference on Communication, Computing and Internet of Things (IC3IoT); February 15-17, 2018; Chennai, Tamil Nadu, India p. 525-530. [CrossRef]
  29. Abedy H, Ahmed F, Bhuiyan M, Islam M, Ali NY, Shamsujjoha M. Leukemia Prediction from Microscopic Images of Human Blood Cell Using HOG Feature Descriptor and Logistic Regression. 2019 Jan 17 Presented at: 2018 16th International Conference on ICT and Knowledge Engineering (ICT&KE); November 21-23, 2018; Bangkok, Thailand p. 1-6. [CrossRef]
  30. Rajpurohit S, Patil S, Choudhary N, Gavasane S, Kosamkar P. Identification of Acute Lymphoblastic Leukemia in Microscopic Blood Image Using Image Processing and Machine Learning Algorithms. 2018 Dec 03 Presented at: 2018 International Conference on Advances in Computing, Communications and Informatics (ICACCI); September 19-22, 2018; Bengaluru, Karnataka, India p. 2359-2363. [CrossRef]
  31. Dharani T, Hariprasath S. Diagnosis of Leukemia and its types Using Digital Image Processing Techniques. 2019 May 30 Presented at: 2018 3rd International Conference on Communication and Electronics Systems (ICCES); October 15-16, 2018; Coimbatore, Tamil Nadu, India p. 275-279. [CrossRef]
  32. Chebouba L, Boughaci D, Guziolowski C. Proteomics Versus Clinical Data and Stochastic Local Search Based Feature Selection for Acute Myeloid Leukemia Patients' Classification. J Med Syst 2018 Jun 04;42(7):129. [CrossRef] [Medline]
  33. Khosla E, Ramesh D. Phase classification of chronic myeloid leukemia using convolution neural networks. 2018 Jun 21 Presented at: 2018 4th International Conference on Recent Advances in Information Technology (RAIT); March 15-17, 2018; Dhanbad, Jharkhand, India p. 1-6. [CrossRef]
  34. Jagadev P, Virani HG. Detection of leukemia and its types using image processing and machine learning. 2018 Feb 22 Presented at: 2017 International Conference on Trends in Electronics and Informatics (ICEI); May 11-12, 2017; Tirunelveli, Tamil Nadu, India p. 522-526. [CrossRef]
  35. Bahra G, Wiese L. Classifying leukemia and gout patients with Neural Networks. In: International Conference on Database and Expert Systems Applications. Cham, Switzerland: Springer; 2018:150-160.
  36. Yu W, Chang J, Yang C, Zhang L, Shen H, Xia Y, et al. Automatic classification of leukocytes using deep neural network. 2018 Jan 11 Presented at: 2017 IEEE 12th International Conference on ASIC (ASICON); October 25-28, 2017; Guiyang, China p. 1041-1044. [CrossRef]
  37. Vincent I, Kwon KR, Lee SH, Moon KS. Acute lymphoid leukemia classification using two-step neural network classifier. 2015 May 11 Presented at: 2015 21st Korea-Japan Joint Workshop on Frontiers of Computer Vision (FCV); January 28-30, 2015; Mokpo, South Korea p. 1-4. [CrossRef]
  38. Chen R, Garapati S, Wu D, Ko S, Falk S, Dierov D. Machine learning based predictive model of 5-year survival in multiple myeloma autologous transplant patients. Blood. (Supplement_1) 2019;134:2156. [CrossRef]
  39. Madanan M. Designing an Artificial Intelligence Model using Machine Learning Algorithms and Applying it to Hematology for the Detection and Classification of Various Stages of Blood Cancer. 2020 Jul Presented at: International Conference on Innovative Technical Advances in Disaster Management (ICITADM); July 15-16, 2020; Kalady, Kerala, India.
  40. Hossain M, Sabik M, Rahman M, Sakiba S, Muzahidul IA, Shatabda S. An effective leukemia prediction technique using supervised machine learning classification algorithm. Singapore City, Singapore: Springer; 2020 Dec 17 Presented at: International Conference on Trends in Computational and Cognitive Engineering; December 17-18, 2020; Dhaka, Bangladesh p. 219-229. [CrossRef]
  41. Licandro R, Reiter M, Diem M, Dworzak M, Schumich A, Kampel M. Application of machine learning for automatic MRD Assessment in Paediatric Acute Myeloid leukaemia. 2018 Presented at: 7th International Conference on Pattern Recognition Applications and Methods; January 16-18, 2018; Madeira, Portugal. [CrossRef]
  42. Lyu M, Radenkovic M, Keskin D, Brusic V. Classification of single cell types during leukemia therapy using artificial neural networks. 2021 Jan 13 Presented at: 2020 IEEE International Conference on Bioinformatics and Biomedicine (BIBM); December 16-19, 2020; Seoul, South Korea p. 1258-1261. [CrossRef]
  43. Patil BS, Mishra S, Junnarkar A. Leukemia diagnosis based on machine learning algorithms. 2021 Jan 01 Presented at: 2020 IEEE International Conference for Innovation in Technology (INOCON). 2020; November 6-8, 2020; Bangluru, Karnataka, India. [CrossRef]
  44. Malkawi A, Al-Assi R, Salameh T, Sheyab B, Alquran H, Alqudah A. White Blood Cells Classification Using Convolutional Neural Network Hybrid System. 2020 Nov 25 Presented at: 2020 IEEE 5th Middle East and Africa Conference on Biomedical Engineering (MECBME); October 27-29, 2020; Amman, Jordan p. 1-5. [CrossRef]
  45. Raina S, Khandelwal A, Gupta S, Leekha A. Blood cells detection using faster-RCNN. 2020 Nov 02 Presented at: 2020 IEEE International Conference on Computing, Power and Communication Technologies (GUCON); October 2-4, 2020; Greater Noida, Uttar Pradesh, India. [CrossRef]
  46. Singh G, Bathla G, Kaur S. A review to detect leukemia cancer in medical images. 2017 Jan 16 Presented at: International Conference on Computing, Communication and Automation (ICCCA); May 5-6, 2017; Greater Noida, NCR, India. [CrossRef]
  47. Tambe R, Mahajan S, Shah U, Agrawal M, Garware B. Towards designing an automated classification of lymphoma subtypes using Deep Neural Networks. 2019 Jan 03 Presented at: ACM India Joint International Conference on Data Science and Management of Data; January 3-5, 2019; Kolkata, West Bengal, India. [CrossRef]
  48. Bhuiyan MNQ, Rahut SK, Tanvir RA, Ripon S. Automatic Acute Lymphoblastic Leukemia Detection and Comparative Analysis from Images. 2019 Presented at: International Conference on Control, Decision and Information Technologies (CoDIT); April 23-26, 2019; Paris, France p. 1144-1149. [CrossRef]
  49. Dey U, Islam M. Genetic Expression Analysis To Detect Type Of Leukemia Using Machine Learning. 2019 Dec 19 Presented at: 2019 1st International Conference on Advances in Science, Engineering and Robotics Technology (ICASERT); May 3-5, 2019; Dhaka, Bangladesh p. 1-6. [CrossRef]
  50. Hassouneh N, Alnemer L, Alsakran J, Rodan A. Predicting Survivability in Leukemia Patients using Deep Learning. 2020 Presented at: 2019 Sixth HCT Information Technology Trends (ITT) Conference; 2019; Ras Al Khaimah, United Arab Emirates. [CrossRef]
  51. Jayashanka R, Wijesinghe C, Weerasinghe A, Pieris D. Machine Learning Approach to Predict the Survival Time of Childhood Acute Lymphoblastic Leukemia Patients. 2018 Presented at: 2018 18th International Conference on Advances in ICT for Emerging Regions (ICTer); September 26-29, 2018; Colombo, Sri Lanka p. 426-432. [CrossRef]
  52. Shurrab A, Maghari A. Blood diseases detection using data mining techniques. 2017 Presented at: 2017 8th International Conference on Information Technology (ICIT); May 17-18, 2017; Amman, Jordan p. 625-631. [CrossRef]
  53. Chulián S, Martínez-Rubio Á, Pérez-García VM, Rosa M, Blázquez Goñi C, Rodríguez Gutiérrez JF, et al. High-Dimensional Analysis of Single-Cell Flow Cytometry Data Predicts Relapse in Childhood Acute Lymphoblastic Leukaemia. Cancers (Basel) 2020 Dec 23;13(1):17 [FREE Full text] [CrossRef] [Medline]
  54. Dasariraju S, Huo M, McCalla S. Detection and Classification of Immature Leukocytes for Diagnosis of Acute Myeloid Leukemia Using Random Forest Algorithm. Bioengineering (Basel) 2020 Oct 01;7(4):120 [FREE Full text] [CrossRef] [Medline]
  55. Agius R, Brieghel C, Andersen MA, Pearson AT, Ledergerber B, Cozzi-Lepri A, et al. Machine learning can identify newly diagnosed patients with CLL at high risk of infection. Nat Commun 2020 Jan 17;11(1):363 [FREE Full text] [CrossRef] [Medline]
  56. Gimenez N, Tripathi R, Giró A, Rosich L, López-Guerra M, López-Oreja I, et al. Systems biology drug screening identifies statins as enhancers of current therapies in chronic lymphocytic leukemia. Sci Rep 2020 Dec 17;10(1):22153 [FREE Full text] [CrossRef] [Medline]
  57. Sun J, Wang L, Liu Q, Tárnok A, Su X. Deep learning-based light scattering microfluidic cytometry for label-free acute lymphocytic leukemia classification. Biomed Opt Express 2020 Nov 01;11(11):6674-6686 [FREE Full text] [CrossRef] [Medline]
  58. Nagata Y, Zhao R, Awada H, Kerr C, Mirzaev I, Kongkiatkamon S, et al. Machine learning demonstrates that somatic mutations imprint invariant morphologic features in myelodysplastic syndromes. Blood 2020 Nov 12;136(20):2249-2262 [FREE Full text] [CrossRef] [Medline]
  59. Kashef AA, Khatibi T, Mehrvar A. Prediction of Cranial Radiotherapy Treatment in Pediatric Acute Lymphoblastic Leukemia Patients Using Machine Learning: A Case Study at MAHAK Hospital. Asian Pac J Cancer Prev 2020 Nov 01;21(11):3211-3219 [FREE Full text] [CrossRef] [Medline]
  60. Zhao M, Mallesh N, Höllein A, Schabath R, Haferlach C, Haferlach T, et al. Hematologist-Level Classification of Mature B-Cell Neoplasm Using Deep Learning on Multiparameter Flow Cytometry Data. Cytometry A 2020 Oct 04;97(10):1073-1080 [FREE Full text] [CrossRef] [Medline]
  61. Bodzas A, Kodytek P, Zidek J. Automated Detection of Acute Lymphoblastic Leukemia From Microscopic Images Based on Human Visual Perception. Front Bioeng Biotechnol 2020;8:1005 [FREE Full text] [CrossRef] [Medline]
  62. Gaidano V, Tenace V, Santoro N, Varvello S, Cignetti A, Prato G, et al. A Clinically Applicable Approach to the Classification of B-Cell Non-Hodgkin Lymphomas with Flow Cytometry and Machine Learning. Cancers (Basel) 2020 Jun 24;12(6):1684 [FREE Full text] [CrossRef] [Medline]
  63. Rodrigues V, Deusdado S. Metalearning approach for leukemia informative genes prioritization. J Integr Bioinform 2020 May 08;17(1):7 [FREE Full text] [CrossRef] [Medline]
  64. Haider RZ, Ujjan IU, Shamsi TS. Cell Population Data-Driven Acute Promyelocytic Leukemia Flagging Through Artificial Neural Network Predictive Modeling. Transl Oncol 2020 Jan;13(1):11-16 [FREE Full text] [CrossRef] [Medline]
  65. Bibi N, Sikandar M, Ud Din I, Almogren A, Ali S. IoMT-Based Automated Detection and Classification of Leukemia Using Deep Learning. J Healthc Eng 2020;2020:6648574 [FREE Full text] [CrossRef] [Medline]
  66. Inbarani H, Azar A, Jothi G. Leukemia Image Segmentation Using a Hybrid Histogram-Based Soft Covering Rough K-Means Clustering Algorithm. Electronics 2020 Jan 19;9(1):188. [CrossRef]
  67. Haferlach T. Advancing leukemia diagnostics: Role of Next Generation Sequencing (NGS) in acute myeloid leukemia. Hematol Rep 2020 Sep 21;12(Suppl 1):8957 [FREE Full text] [CrossRef] [Medline]
  68. Kumar D, Jain N, Khurana A, Mittal S, Satapathy S, Senkerik R, et al. Automatic Detection of White Blood Cancer From Bone Marrow Microscopic Images Using Convolutional Neural Networks. IEEE Access 2020;8:142521-142531. [CrossRef]
  69. Khilji IQ, Saha K, Amin J, Iqbal M. Application of Homomorphic Encryption on Neural Network in Prediction of Acute Lymphoid Leukemia. IJACSA 2020;11(6):e1. [CrossRef]
  70. Vasighizaker A, Sharma A, Dehzangi A. A novel one-class classification approach to accurately predict disease-gene association in acute myeloid leukemia cancer. PLoS One 2019;14(12):e0226115 [FREE Full text] [CrossRef] [Medline]
  71. Togacar M, Ergen B, Sertkaya ME. Subclass Separation of White Blood Cell Images Using Convolutional Neural Network Models. Elektron Elektrotech 2019 Oct 06;25(5):63-68. [CrossRef]
  72. Ahmed N, Yigit A, Isik Z, Alpkocak A. Identification of Leukemia Subtypes from Microscopic Images Using Convolutional Neural Network. Diagnostics (Basel) 2019 Aug 25;9(3):104 [FREE Full text] [CrossRef] [Medline]
  73. Fuse K, Uemura S, Tamura S, Suwabe T, Katagiri T, Tanaka T, et al. Patient-based prediction algorithm of relapse after allo-HSCT for acute Leukemia and its usefulness in the decision-making process using a machine learning approach. Cancer Med 2019 Sep;8(11):5058-5067 [FREE Full text] [CrossRef] [Medline]
  74. Reiter M, Diem M, Schumich A, Maurer-Granofszky M, Karawajew L, Rossi JG, International Berlin-Frankfurt-Münster (iBFM)-FLOW-networkthe AutoFLOW consortium. Automated Flow Cytometric MRD Assessment in Childhood Acute B- Lymphoblastic Leukemia Using Supervised Machine Learning. Cytometry A 2019 Sep 07;95(9):966-975 [FREE Full text] [CrossRef] [Medline]
  75. Gal O, Auslander N, Fan Y, Meerzaman D. Predicting Complete Remission of Acute Myeloid Leukemia: Machine Learning Applied to Gene Expression. Cancer Inform 2019;18:1176935119835544 [FREE Full text] [CrossRef] [Medline]
  76. Castillo D, Galvez JM, Herrera LJ, Rojas F, Valenzuela O, Caba O, et al. Leukemia multiclass assessment and classification from Microarray and RNA-seq technologies integration at gene expression level. PLoS One 2019;14(2):e0212127 [FREE Full text] [CrossRef] [Medline]
  77. Li H, Xu C, Xin B, Zheng C, Zhao Y, Hao K, et al. F-FDG PET/CT Radiomic Analysis with Machine Learning for Identifying Bone Marrow Involvement in the Patients with Suspected Relapsed Acute Leukemia. Theranostics 2019;9(16):4730-4739 [FREE Full text] [CrossRef] [Medline]
  78. Parida L, Haferlach C, Rhrissorrakrai K, Utro F, Levovitz C, Kern W, et al. Dark-matter matters: Discriminating subtle blood cancers using the darkest DNA. PLoS Comput Biol 2019 Aug;15(8):e1007332 [FREE Full text] [CrossRef] [Medline]
  79. Mallesh N, Zhao M, Elsner F, Lueling H, Schabath R, Haferlach C. Knowledge transfer between artificial neural networks for different multicolor flow cytometry protocols improves classification performance for rare B-cell neoplasm subtypes. Blood 2019;134(Supplement_1):2094. [CrossRef]
  80. Pansombut T, Wikaisuksakul S, Khongkraphan K, Phon-On A. Convolutional Neural Networks for Recognition of Lymphoblast Cell Images. Comput Intell Neurosci 2019;2019:7519603 [FREE Full text] [CrossRef] [Medline]
  81. Mosquera Orgueira A, Antelo Rodríguez B, Alonso Vence N, Bendaña López Á, Díaz Arias JÁ, Díaz Varela N, et al. Time to Treatment Prediction in Chronic Lymphocytic Leukemia Based on New Transcriptional Patterns. Front Oncol 2019 Feb 15;9:79 [FREE Full text] [CrossRef] [Medline]
  82. Al-Fahad R, Yeasin M, Glass J, Conklin H, Jacola L, Reddick W. Early Imaging-Based Predictive Modeling of Cognitive Performance Following Therapy for Childhood ALL. IEEE Access 2019;7:146662-146674. [CrossRef]
  83. Syed-Abdul S, Firdani RP, Chung HJ, Uddin M, Hur M, Park JH, et al. Author Correction: Artificial Intelligence based Models for Screening of Hematologic Malignancies using Cell Population Data. Sci Rep 2020 Jun 23;10(1):10475 [FREE Full text] [CrossRef] [Medline]
  84. Deeb SJ, Tyanova S, Hummel M, Schmidt-Supprian M, Cox J, Mann M. Machine Learning-based Classification of Diffuse Large B-cell Lymphoma Patients by Their Protein Expression Profiles. Mol Cell Proteomics 2015 Nov;14(11):2947-2960 [FREE Full text] [CrossRef] [Medline]
  85. Negm AS, Hassan OA, Kandil AH. A decision support system for Acute Leukaemia classification based on digital microscopic images. Alexandria Engineering Journal 2018 Dec;57(4):2319-2332. [CrossRef]
  86. Moshavash Z, Danyali H, Helfroush MS. An Automatic and Robust Decision Support System for Accurate Acute Leukemia Diagnosis from Blood Microscopic Images. J Digit Imaging 2018 Oct;31(5):702-717 [FREE Full text] [CrossRef] [Medline]
  87. Muhsen IN, Shyr D, Sung AD, Hashmi SK. Machine Learning Applications in the Diagnosis of Benign and Malignant Hematological Diseases. Clin Hematol Int 2021 Mar;3(1):13-20 [FREE Full text] [CrossRef] [Medline]
  88. Agarwal N, Agarwal P. Early Stage Detection of Leukemia Using Artificial Intelligence. In: Mohanty SN, Nalinipriya G, Jena OP, Sarkar A, editors. Machine Learning for Healthcare Applications. Hoboken, NJ: Wiley; Apr 12, 2021:215-224.
  89. Bano S, Rao K. Partial Context Similarity of Gene/Proteins in Leukemia Using Context Rank Based Hierarchical Clustering Algorithm. IJECE 2015 Jun 01;5(3):483. [CrossRef]
  90. Ng D, Wu D, Wood B, Fromm J. Computer-aided detection of rare tumor populations in flow cytometry: an example with classic Hodgkin lymphoma. Am J Clin Pathol 2015 Sep;144(3):517-524. [CrossRef] [Medline]
  91. Sirinukunwattana K, Aberdeen A, Theissen H, Sousos N, Psaila B, Mead A, et al. Artificial intelligence-based morphological fingerprinting of megakaryocytes: a new tool for assessing disease in MPN patients. Blood Adv 2020 Jul 28;4(14):3284-3294 [FREE Full text] [CrossRef] [Medline]
  92. Carreras J, Kikuti Y, Miyaoka M, Hiraiwa S, Tomita S, Ikoma H, et al. A Single Gene Expression Set Derived from Artificial Intelligence Predicted the Prognosis of Several Lymphoma Subtypes; and High Immunohistochemical Expression of TNFAIP8 Associated with Poor Prognosis in Diffuse Large B-Cell Lymphoma. AI 2020 Jul 21;1(3):342-360. [CrossRef]
  93. Arai Y, Kondo T, Fuse K, Shibasaki Y, Masuko M, Sugita J. Prediction of acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation using a machine learning algorithm. Blood 2018;132(Supplement 1):68. [CrossRef]
  94. Hamey FK, Göttgens B. Machine learning predicts putative hematopoietic stem cells within large single-cell transcriptomics data sets. Exp Hematol 2019 Oct;78:11-20 [FREE Full text] [CrossRef] [Medline]
  95. Ko B, Wang Y, Li J, Li C, Weng P, Hsu S, et al. Clinically validated machine learning algorithm for detecting residual diseases with multicolor flow cytometry analysis in acute myeloid leukemia and myelodysplastic syndrome. EBioMedicine 2018 Nov;37:91-100 [FREE Full text] [CrossRef] [Medline]
  96. Chen D, Goyal G, Go RS, Parikh SA, Ngufor CG. Improved Interpretability of Machine Learning Model Using Unsupervised Clustering: Predicting Time to First Treatment in Chronic Lymphocytic Leukemia. JCO Clin Cancer Inform 2019 May;3:1-11 [FREE Full text] [CrossRef] [Medline]
  97. Gunčar G, Kukar M, Notar M, Brvar M, Černelč P, Notar M, et al. An application of machine learning to haematological diagnosis. Sci Rep 2018 Jan 11;8(1):411 [FREE Full text] [CrossRef] [Medline]
  98. Elemento O. Toward artificial intelligence driven pathology assessment for hematologic malignancies. Blood Cancer Discovery 2021;2(3):195-197. [CrossRef]
  99. Kaestner L. Artificial intelligence meets hematology. Transfus Apher Sci 2020 Dec;59(6):102986. [CrossRef] [Medline]
  100. Shreve J, Meggendorfer M, Awada H, Mukherjee S, Walter W, Hutter S. A personalized prediction model to risk stratify patients with acute myeloid leukemia (AML) using Artificial Intelligence. Blood 2019;134(Supplement_1):2091. [CrossRef]
  101. Siddiqui N, Klein A, Godara A, Varga C, Buchsbaum R, Hughes M. Supervised machine learning algorithms using patient related factors to predict in-hospital mortality following acute myeloid leukemia therapy. Blood 2019;134(Supplement_1):3435. [CrossRef]
  102. Shouval R, Labopin M, Gorin NC, Bomze D, Houhou M, Blaise D, et al. Individualized prediction of leukemia-free survival after autologous stem cell transplantation in acute myeloid leukemia. Cancer 2019 Oct 15;125(20):3566-3573 [FREE Full text] [CrossRef] [Medline]
  103. Arai Y, Kondo T, Fuse K, Shibasaki Y, Masuko M, Sugita J, et al. Using a machine learning algorithm to predict acute graft-versus-host disease following allogeneic transplantation. Blood Adv 2019 Nov 26;3(22):3626-3634 [FREE Full text] [CrossRef] [Medline]
  104. Morita K, Wang F, Makishima H, Yan Y, Yoshizato T, Yoshida K. Pan-Myeloid Leukemia Analysis: Machine learning-based approach to predict phenotype and clinical outcomes using mutation data. Blood 2018;132(Supplement 1):1801. [CrossRef]
  105. Shouval R, Bonifazi F, Fein J, Boschini C, Oldani E, Labopin M, et al. Validation of the acute leukemia-EBMT score for prediction of mortality following allogeneic stem cell transplantation in a multi-center GITMO cohort. Am J Hematol 2017 May;92(5):429-434 [FREE Full text] [CrossRef] [Medline]
  106. Ananda-Rajah MR, Bergmeir C, Petitjean F, Slavin MA, Thursky KA, Webb GI. Toward Electronic Surveillance of Invasive Mold Diseases in Hematology-Oncology Patients: An Expert System Combining Natural Language Processing of Chest Computed Tomography Reports, Microbiology, and Antifungal Drug Data. JCO Clin Cancer Inform 2017 Nov;1:1-10 [FREE Full text] [CrossRef] [Medline]
  107. Fleming S, Tsai C, Döhner H, Döhner K, Papaemmanuil E. Use of Machine Learning in 2074 Cases of Acute Myeloid Leukemia for Genetic Risk Profiling. Blood 2019;134(Supplement_1):1392. [CrossRef]
  108. Sheikhpour R, Fazli R, Mehrabani S. Gene Identification from Microarray Data for Diagnosis of Acute Myeloid and Lymphoblastic Leukemia Using a Sparse Gene Selection Method. IJPHO 2021 Mar 28;11(2):70-77. [CrossRef]
  109. Patmore R, Smith A, Cathy B, Roman E, Crouch S. Predicting time to treatment in follicular lymphoma using population-based data. Blood 2015;126(23):3912. [CrossRef]
  110. Rezayi S, Mohammadzadeh N, Bouraghi H, Saeedi S, Mohammadpour A. Timely Diagnosis of Acute Lymphoblastic Leukemia Using Artificial Intelligence-Oriented Deep Learning Methods. Comput Intell Neurosci 2021;2021:5478157 [FREE Full text] [CrossRef] [Medline]
  111. Nanaa A, Akkus Z, Lee WY, Pantanowitz L, Salama ME. Machine learning and augmented human intelligence use in histomorphology for haematolymphoid disorders. Pathology 2021 Apr;53(3):400-407. [CrossRef] [Medline]
  112. Boldú L, Merino A, Alférez S, Molina A, Acevedo A, Rodellar J. Automatic recognition of different types of acute leukaemia in peripheral blood by image analysis. J Clin Pathol 2019 Nov 29;72(11):755-761. [CrossRef] [Medline]
  113. Ng D, Zuromski L. Augmented Human Intelligence and Automated Diagnosis in Flow Cytometry for Hematologic Malignancies. Am J Clin Pathol 2021 Mar 15;155(4):597-605. [CrossRef] [Medline]
  114. Sasaki K, Jabbour EJ, Ravandi F, Konopleva M, Borthakur G, Wierda WG, et al. The LEukemia Artificial Intelligence Program (LEAP) in chronic myeloid leukemia in chronic phase: A model to improve patient outcomes. Am J Hematol 2021 Feb 01;96(2):241-250 [FREE Full text] [CrossRef] [Medline]
  115. Fauzi IR, Rustam Z, Wibowo A. Multiclass classification of leukemia cancer data using Fuzzy Support Vector Machine (FSVM) with feature selection using Principal Component Analysis (PCA). J. Phys.: Conf. Ser 2021 Jan 01;1725(1):012012 [FREE Full text] [CrossRef]
  116. Nikitaev VG, Tupitsyn NN, Pronichev AN, Polyakov EV, Dmitrieva VV, Chernysheva OA, et al. Artificial Intelligence Technologies in the Diagnosis of Acute Lymphoblastic Leukemia and Minimal Residual Disease. Biomed Eng 2021 Jan 06;54(5):354-356. [CrossRef]
  117. Mou A, Hasan M, Saha P, Priom N, Saha A. Prediction and Rule Generation for Leukemia using Decision Tree and Association Rule Mining. 2021 Apr 05 Presented at: 2020 11th International Conference on Electrical and Computer Engineering (ICECE); December 17-19, 2020; Dhaka, Bangladesh p. 133-136. [CrossRef]
  118. Li C, Zhu B, Chen J, Huang X. Feature genes predicting the FLT3/ITD mutation in acute myeloid leukemia. Mol Med Rep 2016 Jul;14(1):89-94 [FREE Full text] [CrossRef] [Medline]
  119. Ayyappan V, Chang A, Zhang C, Paidi SK, Bordett R, Liang T, et al. Identification and Staging of B-Cell Acute Lymphoblastic Leukemia Using Quantitative Phase Imaging and Machine Learning. ACS Sens 2020 Oct 23;5(10):3281-3289. [CrossRef] [Medline]
  120. Fathi E, Rezaee MJ, Tavakkoli-Moghaddam R, Alizadeh A, Montazer A. Design of an integrated model for diagnosis and classification of pediatric acute leukemia using machine learning. Proc Inst Mech Eng H 2020 Oct;234(10):1051-1069. [CrossRef] [Medline]
  121. Al-Tahhan FE, Fares ME, Sakr AA, Aladle DA. Accurate automatic detection of acute lymphatic leukemia using a refined simple classification. Microsc Res Tech 2020 Oct;83(10):1178-1189. [CrossRef] [Medline]
  122. Miyoshi H, Sato K, Kabeya Y, Yonezawa S, Nakano H, Takeuchi Y, et al. Deep learning shows the capability of high-level computer-aided diagnosis in malignant lymphoma. Lab Invest 2020 Oct;100(10):1300-1310. [CrossRef] [Medline]
  123. Nazari E, Farzin AH, Aghemiri M, Avan A, Tara M, Tabesh H. Deep Learning for Acute Myeloid Leukemia Diagnosis. J Med Life 2020;13(3):382-387 [FREE Full text] [Medline]
  124. Özyurt F. A fused CNN model for WBC detection with MRMR feature selection and extreme learning machine. Soft Comput 2019 Sep 26;24(11):8163-8172. [CrossRef]
  125. Shafique S, Tehsin S. Acute Lymphoblastic Leukemia Detection and Classification of Its Subtypes Using Pretrained Deep Convolutional Neural Networks. Technol Cancer Res Treat 2018 Jan 01;17:1533033818802789 [FREE Full text] [CrossRef] [Medline]
  126. Madhu G. Computer Vision and machine learning approach for malaria diagnosis in thin blood smears from microscopic blood images. In: Machine Learning for Intelligent Decision Science. Singapore City, Singapore: Springer; Apr 03, 2020:191-209.
  127. Li D, Bledsoe JR, Zeng Y, Liu W, Hu Y, Bi K, et al. A deep learning diagnostic platform for diffuse large B-cell lymphoma with high accuracy across multiple hospitals. Nat Commun 2020 Nov 26;11(1):6004 [FREE Full text] [CrossRef] [Medline]
  128. Sheikh I, Chachoo M. Advanced machine learning for leukaemia detection based on White Blood Cell Segmentation. 2020 Nov 05 Presented at: International Conference on Information, Communication and Computing Technology; May 8, 2020; New Delhi, India p. 195-207. [CrossRef]
  129. Mohammed EA, Mohamed MM, Naugler C, Far BH. Toward leveraging big value from data: chronic lymphocytic leukemia cell classification. Netw Model Anal Health Inform Bioinforma 2017 Feb 18;6(1):e1. [CrossRef]
  130. Su J, Liu S, Song J. A segmentation method based on HMRF for the aided diagnosis of acute myeloid leukemia. Comput Methods Programs Biomed 2017 Dec;152:115-123. [CrossRef] [Medline]
  131. Pan L, Liu G, Lin F, Zhong S, Xia H, Sun X, et al. Machine learning applications for prediction of relapse in childhood acute lymphoblastic leukemia. Sci Rep 2017 Aug 07;7(1):7402 [FREE Full text] [CrossRef] [Medline]
  132. Mohajerani P, Ntziachristos V. Classification of Normal Versus Malignant Cells in B-ALL Microscopic Images Based on a Tiled Convolution Neural Network Approach. In: ISBI 2019 C-NMC Challenge: Classification in Cancer Cell Imaging. Singapore City, Singapore: Springer; 2019:103-111.
  133. Moraes LO, Pedreira CE, Barrena S, Lopez A, Orfao A. A decision-tree approach for the differential diagnosis of chronic lymphoid leukemias and peripheral B-cell lymphomas. Comput Methods Programs Biomed 2019 Sep;178:85-90 [FREE Full text] [CrossRef] [Medline]
  134. Boldú L, Merino A, Acevedo A, Molina A, Rodellar J. A deep learning model (ALNet) for the diagnosis of acute leukaemia lineage using peripheral blood cell images. Comput Methods Programs Biomed 2021 Apr;202:105999. [CrossRef] [Medline]
  135. Coombes C, Abrams Z, Li S, Abruzzo L, Coombes K. Unsupervised machine learning and prognostic factors of survival in chronic lymphocytic leukemia. J Am Med Inform Assoc 2020 Jul 01;27(7):1019-1027 [FREE Full text] [CrossRef] [Medline]
  136. Warnat-Herresthal S, Perrakis K, Taschler B, Becker M, Baßler K, Beyer M, et al. Scalable Prediction of Acute Myeloid Leukemia Using High-Dimensional Machine Learning and Blood Transcriptomics. iScience 2020 Jan 24;23(1):100780 [FREE Full text] [CrossRef] [Medline]
  137. Bachar N, Benbassat D, Brailovsky D, Eshel Y, Glück D, Levner D, et al. An artificial intelligence-assisted diagnostic platform for rapid near-patient hematology. Am J Hematol 2021 Oct 01;96(10):1264-1274. [CrossRef] [Medline]
  138. Bornhauser B, Boutter J, Horvath P, Stanulla M. Image-based RNA interference screening identifies Microenvironmental signals supporting primary acute lymphoblastic leukemia cell survival. Blood 2012;120(21):2348. [CrossRef]
  139. Harkins RA, Chang A, Patel SP, Lee MJ, Goldstein JS, Merdan S, et al. Remaining challenges in predicting patient outcomes for diffuse large B-cell lymphoma. Expert Rev Hematol 2019 Nov;12(11):959-973 [FREE Full text] [CrossRef] [Medline]
  140. Sarno J, Davis KL. Single-cell mass cytometry and machine learning predict relapse in childhood leukemia. Mol Cell Oncol 2018;5(5):e1472057 [FREE Full text] [CrossRef] [Medline]
  141. Tyler E, Jungbluth A, O'Reilly R, Koehne G. WT1-specific T-cell responses in high-risk multiple myeloma patients undergoing allogeneic T cell-depleted hematopoietic stem cell transplantation and donor lymphocyte infusions. Blood 2013 Jan 10;121(2):308-317 [FREE Full text] [CrossRef] [Medline]
  142. Sivapalaratnam S. Artificial intelligence and machine learning in haematology. Br J Haematol 2019 Apr;185(2):207-208. [CrossRef] [Medline]
  143. Mahmood N, Shahid S, Bakhshi T, Riaz S, Ghufran H, Yaqoob M. Identification of significant risks in pediatric acute lymphoblastic leukemia (ALL) through machine learning (ML) approach. Med Biol Eng Comput 2020 Nov;58(11):2631-2640. [CrossRef] [Medline]
  144. Hauser R, Esserman D, Beste L, Ong S, Colomb D, Bhargava A, et al. A Machine Learning Model to Successfully Predict Future Diagnosis of Chronic Myelogenous Leukemia With Retrospective Electronic Health Records Data. Am J Clin Pathol 2021 Nov 08;156(6):1142-1148. [CrossRef] [Medline]
  145. Steyerberg EW. Clinical Prediction Models A Practical Approach to Development, Validation, and Updating. In: Statistics for Biology and Health. Berlin, Germany: Springer; 2020.


AI: artificial intelligence
ALL: acute lymphoblastic leukemia
AML: acute myeloid leukemia
ANN: artificial neural network
AUC: area under the curve
CBC: complete blood count
CLL: chronic lymphocytic leukemia
CLL-TIM: CLL Treatment-Infection Model
CML: chronic myeloid leukemia
CNN: convolutional neural network
CR: complete remission
DL: deep learning
DNN: deep neural network
GOTDP-MP-CNNs: globally optimized transfer deep-learning platform with multiple pretrained CNNs
HSCRKM: histogram-based soft covering rough K-means clustering
IPI: International Prognostic Index
ML: machine learning
RBC: red blood cell
RF: random forest
ROC: receiver operating characteristic
SEER: Surveillance, Epidemiology, and End Results
SESSA: statistically enhanced Salp Swarm Algorithm
SOM: self-organizing map
SVM: support vector machine
WBC: white blood cell


Edited by R Kukafka, G Eysenbach; submitted 16.01.22; peer-reviewed by JA Benítez-Andrades, B Puladi, K Rahmani; comments to author 19.03.22; revised version received 14.05.22; accepted 29.05.22; published 12.07.22

Copyright

©Yousra El Alaoui, Adel Elomri, Marwa Qaraqe, Regina Padmanabhan, Ruba Yasin Taha, Halima El Omri, Abdelfatteh EL Omri, Omar Aboumarzouk. Originally published in the Journal of Medical Internet Research (https://www.jmir.org), 12.07.2022.

This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work, first published in the Journal of Medical Internet Research, is properly cited. The complete bibliographic information, a link to the original publication on https://www.jmir.org/, as well as this copyright and license information must be included.