scholarly journals A Comparative Study of Digital Image Segmentation Algorithms for Acute Myeloid Leukemia M1 White Blood Cells Images

2021 ◽  
Vol 4 (2) ◽  
pp. 101
Author(s):  
Nurcahya Pradana Taufik Prakisya ◽  
Andika Setiawan

Various types of algorithms have been widely used for image segmentation in digital image processing. Every algorithm has features that make it unique to be applied to specific cases. One of the applications of image segmentation is to detect white blood cells. Certain objects such as blood cells must be able to be well segmented because their existence is very crucial to support the accuracy of disease detection related to haematology or the branch of medical science that studies the morphology of blood and blood-forming tissues. Three image segmentation algorithms were compared through this study: Seed Region Growing, Otsu Thresholding and Active Contour Without Edge. Comparative analysis of the three algorithms was done by counting the number of white blood cell objects that were successfully segmented with the actual number of cells that were counted manually. A total of 30 images of blood smears were taken from people suffering from acute myeloid leukemia M1. The average accuracy values from each algorithm were used to determine which image segmentation algorithm is the most suitable for application in the case of white blood cells segmentation. The results showed that Active Contour Without Edge is the most appropriate among the other algorithms

2018 ◽  
Vol 154 ◽  
pp. 01041 ◽  
Author(s):  
Agus Harjoko ◽  
Tri Ratnaningsih ◽  
Esti Suryani ◽  
Wiharto ◽  
Sarngadi Palgunadi ◽  
...  

Acute Myeloid Leukemia (AML) is a type of cancer which attacks white blood cells from myeloid. AML has eight subtypes, namely: M0, M1, M2, M3, M4, M5, M6, and M7. AML subtypes M1, M2 and M3 are affected by the same type of cells, myeloblast, making it needs more detailed analysis to distinguish. To overcome these obstacles, this research is applying digital image processing with Active Contour Without Edge (ACWE) and Momentum Backpropagation artificial neural network for AML subtypes M1, M2 and M3 classification based on the type of the cell. Six features required as training parameters from every cell obtained by using feature extraction. The features are: cell area, perimeter, circularity, nucleus ratio, mean and standard deviation. The results show that ACWE can be used for segmenting white blood cells with 83.789% success percentage of 876 total cell objects. The whole AML slides had been identified according to the cell types predicted number through training with momentum backpropagation. Five times testing calibration with the best parameter generated averages value of 84.754% precision, 75.887% sensitivity, 95.090% specificity and 93.569% accuracy.


2019 ◽  
Vol 18 (14) ◽  
pp. 1936-1951 ◽  
Author(s):  
Raghav Dogra ◽  
Rohit Bhatia ◽  
Ravi Shankar ◽  
Parveen Bansal ◽  
Ravindra K. Rawal

Background: Acute myeloid leukemia is the collective name for different types of leukemias of myeloid origin affecting blood and bone marrow. The overproduction of immature myeloblasts (white blood cells) is the characteristic feature of AML, thus flooding the bone marrow and reducing its capacity to produce normal blood cells. USFDA on August 1, 2017, approved a drug named Enasidenib formerly known as AG-221 which is being marketed under the name Idhifa to treat R/R AML with IDH2 mutation. The present review depicts the broad profile of enasidenib including various aspects of chemistry, preclinical, clinical studies, pharmacokinetics, mode of action and toxicity studies. Methods: Various reports and research articles have been referred to summarize different aspects related to chemistry and pharmacokinetics of enasidenib. Clinical data was collected from various recently published clinical reports including clinical trial outcomes. Result: The various findings of enasidenib revealed that it has been designed to allosterically inhibit mutated IDH2 to treat R/R AML patients. It has also presented good safety and efficacy profile along with 9.3 months overall survival rates of patients in which disease has relapsed. The drug is still under study either in combination or solely to treat hematological malignancies. Molecular modeling studies revealed that enasidenib binds to its target through hydrophobic interaction and hydrogen bonding inside the binding pocket. Enasidenib is found to be associated with certain adverse effects like elevated bilirubin level, diarrhea, differentiation syndrome, decreased potassium and calcium levels, etc. Conclusion: Enasidenib or AG-221was introduced by FDA as an anticancer agent which was developed as a first in class, a selective allosteric inhibitor of the tumor target i.e. IDH2 for Relapsed or Refractory AML. Phase 1/2 clinical trial of Enasidenib resulted in the overall survival rate of 40.3% with CR of 19.3%. Phase III trial on the Enasidenib is still under process along with another trial to test its potency against other cell lines. Edasidenib is associated with certain adverse effects, which can be reduced by investigators by designing its newer derivatives on the basis of SAR studies. Hence, it may come in the light as a potent lead entity for anticancer treatment in the coming years.


2019 ◽  
Author(s):  
Christian Matek ◽  
Simone Schwarz ◽  
Karsten Spiekermann ◽  
Carsten Marr

AbstractReliable recognition of malignant white blood cells is a key step in the diagnosis of hematologic malignancies such as Acute Myeloid Leukemia. Microscopic morphological examination of blood cells is usually performed by trained human examiners, making the process tedious, time-consuming and hard to standardise.We compile an annotated image dataset of over 18,000 white blood cells, use it to train a convolutional neural network for leukocyte classification, and evaluate the network’s performance. The network classifies the most important cell types with high accuracy. It also allows us to decide two clinically relevant questions with human-level performance, namely (i) if a given cell has blast character, and (ii) if it belongs to the cell types normally present in non-pathological blood smears.Our approach holds the potential to be used as a classification aid for examining much larger numbers of cells in a smear than can usually be done by a human expert. This will allow clinicians to recognize malignant cell populations with lower prevalence at an earlier stage of the disease.


2005 ◽  
Vol 114 (2) ◽  
pp. 121-124
Author(s):  
T. Fietz ◽  
R. Arnold ◽  
G. Massenkeil ◽  
K. Rieger ◽  
B. Reufi ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2944-2944 ◽  
Author(s):  
Sonali Lakshminarayanan ◽  
Adam Mendizabal ◽  
Vinod Prasad ◽  
Suhag Parikh ◽  
Paul Szabolcs ◽  
...  

Abstract Busulfan/Melphalan/ATG (Bu/Mel/ATG) have been used as a preparative regimen in children undergoing hematopoetic stem cell transplantation who are too young for or who cannot tolerate total body irradiation (TBI). While successful in a portion of patients, engraftment rates were lower than those seen in a similar patient population receiving a TBI-containing preparative regimen (Wall et al, Biol Blood Marrow Transplant.2005; 11: 637–646). We hypothesized that the addition of fludarabine, a known immunosuppressive agent used in reduced intensity transplants, to this regimen would increase engraftment without additive toxicity. Fourteen pediatric patients with hematological malignancies (acute myeloid leukemia in remission (n=5); acute myeloid leukemia in relapse (n=3), acute lymphoblastic leukemia, infant type in second remission, n=2; juvenile myelomonocytic leukemia, n=1; myelodysplastic syndrome, n=1; biphenotypic leukemia, in first remission, n=1) and non-malignant conditions (infantile myelofibrosis, n=1) were prepared with intravenous fludarabine 25mg/m2 daily × 5days from day-13 to -9, intravenous targeted busulfan 1mg/kg daily × 4 days from day-8 to -5, intravenous melphalan 45mg/m2 daily × 3 days from day-4 to -2 and intravenous equine anti-thymocyte globulin 30mg/kg daily × 3 days from day-3 to -1. Thirteen patients received unrelated donor cord blood cells and one patient received matched related marrow cells. There were 8 male, 8 Caucasian and 4 CMV seropositive patients. The median age was 1.9 years (range 7 months to 21 years). The median pre-cryopreserved total nucleated cell dose per kg for the unrelated cord blood transplant patients was 11.5 × 107/kg (range, 3.4–32.5) while the median post-thaw total nucleated cell dose per kg was 7.3 × 107/kg (range, 2.5–27.6). The median infused CD34 cells/kg was 0.2 × 105/kg (range, 0.1–0.5) and the median CD3 cells/kg was 19.6 × 106 × kg (range, 6.9–31.0). Nine patients received 4/6 HLA-matched cord blood cells, 3 received 5/6 and 2 patients received 6/6 matched grafts. The addition of fludarabine did not increase or result in any unexpected toxicity. The cumulative incidence (CINC) and median time to neutrophil and platelet engraftment, acute graft versus host disease and overall survival are shown in the table below. These data were compared to a previous published study. Engraftment of neutrophils was superior in the fludarabine/Busulfan/Melphalan/ATG group. Overall survival also improved. While these data of patients are small, these results suggest that the addition of fludarabine to a Busulfan/Melphalan/ATG regimen is safe and may facilitate engraftment with improved survival. Comparison of outcome between Bu/Mel/ATG and Flu/Bu/Mel/ATG CINC of neutrophil engraftment at day 42 CINC of platelet engraftment 50K at day 100 CINC of GrII-IV aGVHD at 100days Overall survival at 12 months Relapse-free survival at 12 months Bu/Mel/ATG 59% (95%CI,44%–78%) 40% (95%CI,31%–69%) 41% (95%CI,25%–56%) 47% (95%CI,30%–64%) 34.4% (95%CI,18.8%–50.6%) Flu/Bu/Mel/ATG 71.4% (95%CI,46%–96.8%) 41.5% (95%CI,10.6%–72.5%) 42.9% (95%CI,29%–56.7%) 55% (95%CI, 24.6%–85.1%) 43.9% (95%CI,13%–74.8%)


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1283-1283 ◽  
Author(s):  
Maaike Luesink ◽  
Jeannet Nigten ◽  
Ruth H.J.N. Knops ◽  
Theo J.M. de Witte ◽  
Bert A. van der Reijden ◽  
...  

Abstract Abstract 1283 Poster Board I-305 Wilms' tumor 1 (WT1) and GATA binding protein 2 (GATA2) transcription factors are highly expressed in hematopoietic stem cells and progenitors. Differentiation of precursor blood cells towards mature blood cells is accompanied by rapid downregulation of both transcription factors. Overexpression of WT1 has been observed in the majority of acute myeloid leukemia (AML) cases. Furthermore, in 10-15% of the AML cases mutations in the WT1 gene occur, which have been correlated with poor prognosis. Aberrant expression of GATA2 in AML has been described as well, but no mutations in this gene have been reported in AML so far. How the (aberrant) expression of WT1 and GATA2 is controlled is not completely clear. A regulatory role for microRNAs (miRNAs) has been described for several transcription factors which regulate hematopoiesis. MiRNAs negatively regulate gene expression by translational repression or degradation of target messenger RNAs (mRNAs). In the present study we investigated the interplay between miRNAs and transcription factors that are involved in myeloid development and malignant transformation towards AML. We studied the expression of 158 miRNAs in the APL cell line NB4 during induction of granulocytic differentiation with all-trans retinoic acid (ATRA). Quantitative PCR specific for mature miRNAs was performed (Applied Biosystems). Twenty out of 158 miRNAs were more than 10-fold upregulated upon differentiation induction with ATRA. MiR-132 and miR212, which are derived from the same pri-miRNA transcript, were most strongly upregulated during ATRA-induced granulocytic differentiation (1200- and 350-fold respectively at 96 hours after ATRA-stimulation). In vitro ATRA-induction of primary APL cells also resulted in upregulation of miR-132 and miR-212. Computational target prediction algorithms were used to identify transcription factors which may be targeted by miR-132 and miR-212. Subsequently, the expression pattern of the predicted targets was determined experimentally in NB4 cells before and after differentiation induction with ATRA using microarray-based mRNA profiling (Affymetrix). In addition, further verification of target gene expression during ATRA-induced differentiation was performed using quantitative PCR. The transcription factors WT1 and GATA2 were predicted as targets of miR-132 and miR-212 by two out of four different prediction programs that were used. Both transcription factors contained putative binding sites for miR-132 and miR-212 in their 3'UTR. When tested on microarray and by quantitative PCR, the expression of WT1 and GATA2 was indeed strongly downregulated during ATRA-induced granulocytic differentiation of NB4 cells (65- and 165-fold respectively at 96 hours after ATRA stimulation) as well as in primary leukemia cells derived from APL patients (30- and 10-fold respectively at 48 hours after ATRA-stimulation). During ATRA-induced differentiation the expression levels of WT1 were positively correlated with the expression levels of GATA2. In addition, WT1 expression was also strongly correlated with GATA2 expression in a cohort of 27 pre-treatment AML cases as well as in 7 healthy controls, suggesting that these genes might be co-regulated to a large extent. To directly prove that WT1 and GATA2 are indeed targeted by miR-132 and miR-212, we are currently performing lentiviral-based overexpression studies of both miRNAs to determine the effect on endogenous WT1 and GATA2 mRNA expression. MicroRNAs which target WT1 and GATA2 may be valuable tools in controlling the aberrant expression of WT1 and GATA2 observed in AML. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4818-4818
Author(s):  
Yanwen Zheng ◽  
Zhengwei Mao ◽  
Bin Yin

Abstract Abstract 4818 Acute myeloid leukemia (AML) is a detrimental disease with difficult diagnosis and treatment. Understanding the biology of AML at the molecular and cellular levels would be essential to successful management of the disease. However, the notoriously known difficulty in manipulation of leukemia cells has long hindered the dissection of AML pathogenesis. The advent of CdSe/ZnS quantum dots (QDs) represents an important advancement in the research field of nanotechnology, which have recently also been applied for imaging of live cells. Here, we have introduced a non-genetic approach of marking blood cells, by taking advantage of QD technology. We compared QDs complexed with different vehicles, including a peptide Tat (QDs-Tat), cationic polymer Turbofect (QDs-Tf) and liposome Lipofectamine 2000 (QDs-Lip), in their abilities to mark cells. QDs-Tat showed the highest efficiency in delivery into hematopoietic cells, among the three vehicles. We then examined QDs-Tat labelling of leukemia cell lines, and found that QDs-Tat could label 293T, bone marrow (BM) cells, THP-1, MEG-01 and HL-60 with a decreasing efficiency. The efficiency of QDs-Tat delivery was dependent on the concentration of QDs-Tat applied, but not the length of incubation time. In addition, more uniform intracellular distributions of QDs in 293T and leukemia cells were obtained with QDs-Tat, compared with the granule-like formation obtained with QDs-Lip. Clearly, QD fluorescence was sharp and tolerant to repetitive photo excitations, and could be detected in 293T for up to one week following labelling. In summary, our results suggest that QDs have provided a photostable, non-genetic and transient approach that labels normal and malignant hematopoietic cells in a cell type-, vehicle-, and QD concentration-dependent manner. We expect for potentially wide applications of QDs as an easy and fast tool assisting investigations of various types of blood cells in the near future. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 941-941
Author(s):  
Roland B. Walter ◽  
Megan Othus ◽  
Elisabeth M. Paietta ◽  
Janis Racevskis ◽  
Hugo F Fernandez ◽  
...  

Abstract Background: Therapeutic resistance remains the primary challenge in adult acute myeloid leukemia (AML). Genetic profiling can refine the prediction of outcome across populations of AML patients and has enabled the development of integrated mutational/cytogenetic risk schemas that can separate patients with cytogenetically defined intermediate-risk AML into three subgroups with markedly different outcomes. Here, we investigated to what degree the prediction of therapeutic resistance and survival can be improved for individual patients by inclusion of data from genetic profiling. Patients and Methods: We used data on adults aged 17-60 years with newly diagnosed AML who received treatment on a recent phase 3 trial from the Eastern Cooperative Study Group that investigated the value of escalated doses of daunorubicin during induction (E1900; NCT00049517). We used several criteria for the definition of therapeutic resistance: (a) failure to attain complete remission (CR) despite surviving at least 28 days from beginning induction therapy (“primary refractory”); (b) primary refractory or relapse-free survival (RFS) ²3 months; (c) primary refractory or RFS ²6 months; and (d) primary refractory or RFS ²12 months. We used logistic regression analyses to assess the relationship between individual covariates and measures of resistance and overall survival (OS): age, gender, white blood cell (WBC) count, platelet count, bone marrow blast percentage, disease type (primary vs. secondary), cytogenetic risk, and mutational status in the following genes: ASXL1, CEBPA, DNMT3A, FLT3, IDH1, IDH2, KIT, KRAS, MLL, NPM1, NRAS, PHF6, RUNX1, TET2, TP53, and WT1. The integrated mutational/cytogenetic risk schema was used as established by Patel et al. (NEJM 2012;366:1079-1089). We then used the area under the receiver operator characteristic curve (AUC) to quantify a multivariate modelÕs ability to predict therapeutic resistance; in this approach, an AUC of 1 indicates perfect prediction while an AUC of 0.5 indicates no prediction; AUC values of 0.6-0.7, 0.7-0.8, and 0.8-0.9 are commonly considered as poor, fair, and good, respectively. Results: 298 patients surviving at least 28 days had data on all covariates and were included. 201 (67.4%) of these achieved CR and 97 (32.6%) were primary refractory; 103/297 patients (34.7%) with sufficient follow-up time were either primary refractory or had a RFS of ²3 months, 115/296 patients (38.9%) with sufficient follow-up time were either primary refractory or had a RFS of ²6 months, and 153/295 patients (51.9%) with sufficient follow-up time were primary refractory or had a RFS of ²12 months. The integrated mutational/cytogenetic risk schema was the most important individual predictor of resistance (AUCs ranging between 0.64 and 0.69 across the several definitions of resistance) and survival (AUC of 0.65), followed by cytogenetic risk and FLT3/NPM1 status (AUCs ranging between 0.59 and 0.64). Bootstrap-corrected multivariate models yielded AUCs of 0.76-0.79 for the prediction of primary refractoriness or primary refractoriness/RFS of 3 months or less, 6 months or less, or 12 months or less, respectively, and an AUC of 0.72 for the prediction of OS. Removal of information on FLT3/NPM1 status or mutational data from other profiled genes decreased the AUC to about the same degree each, yielding AUCs of 0.66-0.72 for simpler models including cytogenetic risk and other basic information (age, gender, performance status, white blood cells, platelet counts, marrow blast percentage; see table). Conclusion: Genetic profiling increases the accuracy of multivariate models predicting therapeutic resistance or survival in adult AML. Nevertheless, even with inclusion of such data, our ability to predict these outcomes based on pre-treatment information remains relatively limited. This finding would argue for the integration of treatment response measures (e.g. minimal residual disease) to optimize prediction of resistance. Table Parameter No CR No CR or RFS 3 months or less No CR or RFS 6 months or less No CR or RFS 12 months or less OS Basic model 0.60 0.60 0.63 0.63 0.59 Basic model + Cytogenetic risk 0.68 0.68 0.71 0.72 0.66 Basic model + Integrated mutational/ cytogenetic risk schema 0.68 0.68 0.71 0.74 0.68 Basic model + Cytogenetic risk + NPM1, FLT3/ITD 0.72 0.71 0.74 0.75 0.69 Basic model + Cytogenetic risk + NPM1, FLT3/ITD + Other mutations 0.76 0.76 0.78 0.79 0.72 Disclosures Levine: Novartis: Consultancy, Grant support Other.


2020 ◽  
Vol 40 (6) ◽  
Author(s):  
Xin Zhu ◽  
Qian Zhao ◽  
Xiaoyu Su ◽  
Jinming Ke ◽  
Yunyun Yi ◽  
...  

Abstract The identification of effective signatures is crucial to predict the prognosis of acute myeloid leukemia (AML). The investigation aimed to identify a new signature for AML prognostic prediction by using the three-gene expression (octamer-binding transcription factor 4 (OCT4), POU domain type 5 transcription factor 1B (POU5F1B) and B-cell-specific Moloney murine leukemia virus integration site-1 pseudogene 1 (BMI1P1). The expressions of genes were obtained from our previous study. Only the specimens in which three genes were all expressed were included in this research. A three-gene signature was constructed by the multivariate Cox regression analyses to divide patients into high-risk and low-risk groups. Receiver operating characteristic (ROC) analysis of the three-gene signature (area under ROC curve (AUC) = 0.901, 95% CI: 0.821–0.981, P<0.001) indicated that it was a more valuable signature for distinguishing between patients and controls than any of the three genes. Moreover, white blood cells (WBCs, P=0.004), platelets (PLTs, P=0.017), percentage of blasts in bone marrow (BM) (P=0.011) and complete remission (CR, P=0.027) had significant differences between two groups. Furthermore, high-risk group had shorter leukemia-free survival (LFS) and overall survival (OS) than low-risk group (P=0.026; P=0.006), and the three-gene signature was a prognostic factor. Our three-gene signature for prognosis prediction in AML may serve as a prognostic biomarker.


Sign in / Sign up

Export Citation Format

Share Document