ChapterPDF Available

Brain Tumor Classification Using Convolutional Neural Network

Authors:

Abstract

Misdiagnosis of brain tumor types will prevent effective response to medical intervention and decrease the chance of survival among patients. One conventional method to differentiate brain tumors is by inspecting the MRI images of the patient’s brain. For large amount of data and different specific types of brain tumors, this method is time consuming and prone to human errors. In this study, we attempted to train a Convolutional Neural Network (CNN) to recognize the three most common types of brain tumors, i.e. the Glioma, Meningioma, and Pituitary. We implemented the simplest possible architecture of CNN; i.e. one each of convolution, max-pooling, and flattening layers, followed by a full connection from one hidden layer. The CNN was trained on a brain tumor dataset consisting of 3064 T-1 weighted CE-MRI images publicly available via figshare Cheng (Brain Tumor Dataset, 2017 [1]). Using our simple architecture and without any prior region-based segmentation, we could achieve a training accuracy of 98.51% and validation accuracy of 84.19% at best. These figures are comparable to the performance of more complicated region-based segmentation algorithms, which accuracies ranged between 71.39 and 94.68% on identical dataset Cheng (Brain Tumor Dataset, 2017 [1], Cheng et al. (PLoS One 11, 2017 [2]).
Brain Tumor Classification Using
Convolutional Neural Network
Nyoman Abiwinanda, Muhammad Hanif, S. Tafwida Hesaputra,
Astri Handayani, and Tati Rajab Mengko
Abstract
Misdiagnosis of brain tumor types will prevent effective
response to medical intervention and decrease the chance
of survival among patients. One conventional method to
differentiate brain tumors is by inspecting the MRI images
of the patients brain. For large amount of data and
different specic types of brain tumors, this method is
time consuming and prone to human errors. In this study,
we attempted to train a Convolutional Neural Network
(CNN) to recognize the three most common types of brain
tumors, i.e. the Glioma, Meningioma, and Pituitary. We
implemented the simplest possible architecture of CNN;
i.e. one each of convolution, max-pooling, and attening
layers, followed by a full connection from one hidden
layer. The CNN was trained on a brain tumor dataset
consisting of 3064 T-1 weighted CE-MRI images pub-
licly available via gshare Cheng (Brain Tumor Dataset,
2017 [1]). Using our simple architecture and without any
prior region-based segmentation, we could achieve a
training accuracy of 98.51% and validation accuracy of
84.19% at best. These gures are comparable to the
performance of more complicated region-based segmen-
tation algorithms, which accuracies ranged between 71.39
and 94.68% on identical dataset Cheng (Brain Tumor
Dataset, 2017 [1], Cheng et al. (PLoS One 11, 2017 [2]).
Keywords
Training loss Training accuracy Validation loss
Validation accuracy Overtting
1 Introduction
On 2016, brain tumor was the leading cause of
cancer-related death in children (ages 014) in the United
States and ranked above Leukemia [3]. Brain and CNS
tumors are also the third most common cancer occurring
among teenager and adolescents (ages 1539) [4]. Different
types of brain tumors require different medical interventions.
In conventional computer-aided diagnosis systems, the
tumor mass itself has to be identied and segmented before
it can be classied into different types. Upon tumor mass
segmentation, the segmented region is then subjected to
feature extraction and classication.
Recent studies of identication and segmentation of brain
tumor [5,6] found no universal system for accurate tumor
detection system regardless of its location, shape, and
intensity [6]. There are numerous proposed algorithms in
recent studies for feature extraction and classication of
brain tumors. Grey-level co-occurrence matrix (GLCM) [7
9] is commonly used for extraction of low-level features.
Several other feature extraction algorithms which attempt to
handle the complex texture of brain tumor are Neural Net-
work [9,10], Bag-of-Words (BoW) [2,8], and Fisher Vector
[2]. One recent study showed that by using a combination of
adaptive spatial pooling and sher vector algorithm, brain
tumor classication into Glioma, Meningioma, and Pituitary
can be achieved with 71.3994.68% accuracy [2].
Conventional brain tumor classication methods com-
monly involve region-based tumor segmentation prior to
feature extraction and classication. In this paper, we propose
an automatic brain tumor segmentation/classication method
based on Convolutional Neural Networks. CNN consists of a
convolutional network to perform automatic segmentation
N. Abiwinanda (&)M. Hanif S. T. Hesaputra A. Handayani
T. R. Mengko
Bandung Institute of Technology, Bandung, West Java 40134,
Indonesia
e-mail: abiwinanda@outlook.com
M. Hanif
e-mail: mhaniffarhat@gmail.com
S. T. Hesaputra
e-mail: tafwidahesaputra@gmail.com
A. Handayani
e-mail: a.handayani@stei.itb.ac.id
T. R. Mengko
e-mail: tmengko@stei.itb.ac.id
©Springer Nature Singapore Pte Ltd. 2019
L. Lhotska et al. (eds.), World Congress on Medical Physics and Biomedical Engineering 2018,
IFMBE Proceedings 68/1, https://doi.org/10.1007/978-981-10-9035-6_33
183
and feature extraction, followed by a conventional neural
network to perform classication task. The well-known basic
architecture of CNN involves a Rectied Linear Unit (ReLu),
a convolution, and a pooling layer [11]. In contrast to con-
ventional methods which require prior segmentation of tumor
mass, our CNN approach does not involve region-based
pre-processing step. We validate our algorithm on the same
dataset which was used in previous publications [1,2].
2 Previous Work
Various methodologies have been developed in the past years
to recognize brain tumor in MRI images. These methods are
ranging from classical image processing to neural network
based machine learning approach. Jun Cheng et al. [2]
developed a tumor classication method that consists of two
phases: ofine database building and online retrieval. In the
ofine database phase, the brain tumor images are processed in
sequential steps. The steps are consisted of tumor segmenta-
tion, feature extraction, and distance metric learning. In the
online learning, the input brain image will be processed sim-
ilarly and compare the extracted feature with the learned dis-
tanced metrics which are stored in the online database. This
method does not use neural network approach but could
achieve a classication accuracy of 94.68%. On the other
hand, Gawande and Mendre [12] used Deep Neural Network
using autoencoders in order to classify the brain tumor. Image
segmentation and feature extraction had been implemented on
the image before it was processed by DNN layers. The texture
and intensity based features of the image were extracted with
help of Gray Level Co-occurence Matrix (GLCM) and Dis-
crete Wavelet Transform (DWT). In the nal step, DNN layers
which consist of two autoencoders and one softmax layer were
performed for classication. Furthermore, Pereira et al. [13]
also exploring the used of Convolutional Neural Networks
(CNN) with small 3 3 kernels in order to get to the deeper
architecture and avoid the overtting. They also investigated
the use of intensity normalization as the pre-processing step
before getting into the CNN layers. In this study, inspired by
those works, we investigate and explore the implementation of
deep CNN on classifying several brain tumor type diagnosis
problems in order to get better accuracy result.
3 Method
3.1 Convolutional Neural Network
CNN convolution layer is a network where an image will be
convolved with lters to produce feature maps. This feature
maps will be forwarded to the next convolution layer to
receive or extract another higher level features from the input
image. Between convolution layers, non-linearity functions
and down sampling operation is used to add non-linearity and
reduced the dimensionality of the image respectively. Max-
pooling usually used as the down sampling operation as it
reduced the dimension while preserving the dominant feature
in the feature maps. Just after the last convolution layer or
before the rst layer of the neural network, attening layer
exist to vectorize the feature maps. In the neural network or
classication phase, the atten input vector will be forwarded
into the network to produce a number at each output neurons.
This number tells how much an input vector is classied as a
certain class. Usually a softmax activation function is used at
the output layer to normalize the output sum such that all
numbers at the output neuron will add up to one.
In the training phase, CNN use a learning or optimizer
algorithm to update the lters at the convolution layers and
weights at the neural network or fully connected layer. The
learning algorithm takes a classication error or loss as an
input and back propagates the error into the network to
update the lters and the weights.
3.2 CNN Architecture
In this paper, we use ve different architectures to test the
accuracy of brain tumor classication. There are already well
dened CNN architecture such as AlexNet [14], VGG16
[15], and ResNet [16] but the architecture that are imple-
mented in this paper are much simpler that the one men-
tioned. The CNN architecture that are implemented in this
paper are summarize in the Fig. 1ae.
3.3 Hyper-parameters Optimization
Hyper-parameter is a parameter in deep learning process
whose value can be set and tuned before the learning pro-
cess. This parameter will determine the algorithm to be used
in the learning process. Different model training algorithms
need different hyper-parameters that also affecting the result
of the learning process too. Hyper-parameter optimizer has
to be chosen and tuned so that the classier will have the
most optimal way to solve the problem.
In this study, we will use adamoptimizer in the learning
process which is a method for stochastic optimization by
utilizing the stochastic gradient descent principle. Adam
optimizer which stands for adaptive moment estimation is
chosen because of its advantage that can handle sparse
gradients on noisy problems.
184 N. Abiwinanda et al.
4 Data
In this paper, our CNN is trained using 3064 T-1 weighted
CE-MRI of brain tumor images. The dataset is provided by
Jun Cheng and was previously used in his paper [1,2]. This
dataset consists of 708 images with glioma, 1426 images with
meningioma, and 930 images with pituitary tumors. In our
training phase, we equalize the amount of images that are
used to train the CNN for each class or type of tumors. Out of
all available images, we only used 700 images from each
class where 500 of those images were used for training phase
and the other 200 images were used for validation phase. The
dataset was originally provided in matlab.mat format where
each le stores a struct containing a label which specify the
type of tumor for a particular brain image, patient ID, image
data in 512 512 uint16 format, vector storing the
coordinates of discrete points on tumor border, and a binary
mask image with 1 s indicating tumor region. In our paper we
only make use the label and image data in the.mat les
therefore our brain tumor classier is a simple CNN network
which only takes image as an input. Figure 2ac represent
example of the dataset from each of the classes.
5 Result
In our experiment, the hyperparameter at each layer such as
number and size of lters in the convolution layers, size of
maxpooling kernel, number of neurons in the fully con-
nected layers are held xed. Only the depth of the archi-
tecture are varied between different architectures. The
architecture and hyperparameter that are used are served in
Table 1.
Fig. 1 aeThe proposed
Convolutional Neural Network
Architecture 15
(a) (b) (c)
Fig. 2 aGlioma, bMeningioma,
cPituitary (each labeled in green)
Brain Tumor Classification Using Convolutional Neural Network 185
Sizes of the input images that are forwarded into the
network are 64 64. The original images are in the size of
512 512. This reduction is performed because of com-
putational cost reason. All architectures are compiled with-
out a GPU therefore to speed up the training phase smaller
image size is used.
All convolution layers in the architectures use 32 lters of
size 3 3. We use ReLu as our activation function as it
already the standard activation function used in image
classication task. The size of the maxpool kernel is 2 2
and all the fully connected layer (called densein keras) use
64 neurons.
Finally, there are 3 neurons in the output layer since we
try to classify an image with three types of brain tumors
(glioma, meningioma, and pituitary). The activation func-
tions that are used at the output layer are softmax so that all
three output neurons are summed up to one.
Based on Table 1, each of architecture produces different
numbers of params and features depending on the depth of
the convolution layer and the fully connected network. The
Table 1 Architecture 15
Architecture 1 Architecture 2
Architecture 3 Architecture 4
Architecture 5
186 N. Abiwinanda et al.
architecture with deeper layers of convolution will have
fewer numbers of the trainable params.
Implementation of the above architectures will produce
four parameter values that will describe the success of the
classier model in classifying the input image. The four
parameter values are loss and accuracy from the training set
and validation set. Accuracy is dened as the percentage of
the correct guesses by the classier either for the training set
input or the validation set input. Loss is dened as feasible
error that represents the price paid for inaccuracy of pre-
dictions in classication problem. We use cross-entropy
method for loss calculation. The cross-entropy loss calcu-
lation can be represented in the mathematical form [17]
below:
Hðy;b
yÞ¼X
i
yilog 1
b
yi
¼X
i
yilog b
yið1Þ
yirepresents the result of the classier output from class i.
While b
yirepresents the expected output from class i. The
classication accuracy of each architecture are presented in
Figs. 3,4,5,6and 7.
Based on Figs. 3,4,5,6and 7, the values of loss and
accuracy vary according to the implementation of the
architecture. The classier model is said to be good tif
the accuracy of training set and validation set tend to
increase for every epoch of training. However, if the accu-
racy of validation set tends to decrease while the accuracy of
training set increases, then the classier model is estimated
to have overtting. Overtting happens when the model
learns the detail and noise in the training data thus reducing
its ability to generalize other datasets well [18].
By looking at the result of each architectures, we could
see that all architectures validation loss shows an increasing
trend with respect to the number of epoch except for
architecture 2. This indicates that architecture 2 is the best
architecture out of the ve architectures at generalizing the
brain tumor images. The decreasing pattern in the validation
loss indicates that using the available training images,
architecture 2 could classify the unknown images in the
Fig. 3 Accuracy and loss of architecture 1
Fig. 4 Accuracy and loss of architecture 2
Fig. 5 Accuracy and loss of architecture 3
Fig. 6 Accuracy and loss of architecture 4
Brain Tumor Classification Using Convolutional Neural Network 187
validation set with medium performance. We conclude a
medium performance since the validation loss does not show
a perfect decreasing pattern as the number of epoch increase.
In the last epoch, architecture 2 could achieve a validation
accuracy of 84.19%.
Based on the best architectural choice on the previous
part, we try to vary the number of the lter in the convo-
lution layer into 64 lters and 128 lters we called it as
architecture 6 and architecture 7 respectively. In this
experiment, we will identify the effect of the lter numbers
in the convolution layer on the accuracy of the validation set.
The architecture and hyper parameters that are used are
served in the Table 2.
After implementation of those architectures, the classi-
cation accuracy of those architectures are presented in
Figs. 8and 9.
From the result, the architecture that has the highest
validation accuracy is still architecture 2 with 32 lters in the
convolution layers. Although an increase in the number of
lters in the convolution layer does not necessarily con-
tribute to better CNN performance, in our case the number of
lter in a convolution layer does inuence the accuracy of
the classier. We stick or recommend the use of architecture
2 to classify a brain tumor since it has the highest validation
accuracy.
Fig. 7 Accuracy and loss of architecture 5
Table 2 Architecture 67
Architecture 6 Architecture 7
Fig. 8 Accuracy and loss of architecture 6 Fig. 9 Accuracy and loss of architecture 7
188 N. Abiwinanda et al.
6 Conclusion
In this paper, we introduced CNN to automatically classify
the three most common types of brain tumor, i.e. the Glioma,
Meningioma, and Pituitary; without requiring region-based
pre-processing steps. We identied an optimal CNN archi-
tecture (architecture 2) consisting of 2 layers of convolution,
activation (ReLu), and maxpool, followed by one hidden
layer of 64 neurons. Architecture 2 is the only architecture
that show a consistently decreasing pattern in the validation
loss as the number of epoch increases, leading to the highest
validation accuracy out of all ve architectures. The training
and validation accuracies of architecture 2 at best is 98.51%
and 84.19%, respectively. These gures, although somewhat
lower, are still comparable to the accuracies of conventional
algorithms with region-based pre-processing, which per-
formed at 71.3994.68% [1,2]. For future work, we con-
sider to include color balancing step into our CNN, to
improve classication accuracy of textured brain MRI pixels
[19]. Our algorithm may be implemented in as a simple
supportive tool for medical doctor in classifying brain tumor.
Disclosure The authors declare that they have no conict of interest.
References
1. Jun Cheng. (2017). Brain Tumor Dataset (Version 5). Retrieved
from https://doi.org/10.6084/m9.gshare.1512427.v5
2. Cheng, J., Huang, W., Cao, S., et al. (2015). Classication via
Tumor Region Augmentation and Partition. PLoS One,10(10).
3. Varade, A. A., Ingle, K. S. (2017). Brain MRI Classication
Using PNN and Segmentation Using K Means Clustering,
International Journal of Advanced Research in Electrical, Elec-
tronics and Instrumentation Engineering,6(5), 61816188.
4. Cheng, J., Yang, W., Huang, M., et al. (2016). Retrieval of Brain
Tumors by Adaptive Spatial Pooling and Fisher Vector Represen-
tation. PLoS One,11(6).
5. Lavanyadevi, R., Machakowsalya, M., Nivehitha, J., et al. (2017),
Brain tumor classication and segmentation in MRI images using
PNN, IEEE Xplore.
6. Siegel, R. L., Miller, K. D., Jemal, A. (2016). Cancer statistics,
2016. CA: A Cancer Journal for Clinicans,66(1), 0730.
7. Quinn, T., Ostrom, M.A., Gittleman, H., et al. (2016). American
Brain Tumor Association Adolescent and Young Adult Primary
Brain and Central Nervous System Tumors Diagnosed in the
United States in 20082012. Neuro-Oncology,18(1), i1i50.
8. Gordillo, N., Montseney, E., Sobrevilla, P. (2013). State of the art
survey on MRI brain tumor segmentation. Magnetic Resonance
Imaging,31(8), 14261438.
9. Kaur, H., Sharma, R. (2016). A Survey on Techniques for Brain
Tumor Segmentation from MRI. IOSR Journal of Electronics and
Communication Engineering,11(5), 0105.
10. Hasan, M. H., Meziane, F., et al. (2016). Segmentation of Brain
Tumors in MRI Images Using Three-Dimensional Active Contour
without Edge. Symmetry,8(11), 132.
11. Wu, J. (2017). Introduction to Convolutional Neural Networks.
National Key Lab for Novel Software Technology Nanjing
University, China.
12. He, K., Zhang, X., Ren, S., et al. (2015). Deep residual learning for
Image Recognition. CVPR, 770778.
13. Browniee, J. (2016, March 21). Overtting and Undertting With
Machine Learning Algorithms. Retrieved from https://
machinelearningmastery.com/overtting-and-undertting-with-
machine-learning-algorithms/.
14. Zacharaki, E. I., Wang, Sumei., Chawla, S., et al. (2009).
Classication of Brain Tumor Type and Grade Using MRI Texture
and Shape in a Machine Learning Scheme.Magnetic Resonance in
Medicine, 62(6), 16091618.
15. Kingma, D. P., Ba, J. (2014). Adam: A Method for Stochastic
Optimization, arXiv preprint arXiv:1412.6980.
16. Gawande, S. S., Mendre, V. (2017). Brain Tumor Diagnosis Using
Deep Neural Network (DNN). International Journal of Advanced
Research in Electrical, Electronics and Instrumentation Engineer-
ing, 5(5).
17. Simonyan, K., Zisserman, A. (2015). Very Deep Convolution
Networks for Large-Scale Image Recognition, ICLR 2015.
18. Pereira, S., Pinto, A., Alves, V., et al. (2016). Brain Tumor
Segmentation Using Convolutional Neural Networks in MRI
Images. IEEE Transactions on Medical Imaging, 35(5), 12401251.
19. DiPietro, Rob. (2016, May 2). A Friendly Introduction to
Cross-Entropy Loss. Retrieved from https://rdipietro.github.io/
friendly-intro-to-cross-entropy-loss/.
Brain Tumor Classification Using Convolutional Neural Network 189
... Among these, there have been many which used the dataset published by Jun Cheng [7]. In [35], the authors proposed 5 convolutional neural network architectures, which were much simpler than other state-of-the-art methods while still achieving satisfactory results. The highest accuracy that the authors achieved was 84.19%. ...
... First, the dataset contains more than 3,000 images, which can be sufficient to train deep learning-based solutions that need huge amounts of data. It has also been tested and verified in the clinical setting and was used as the foundation of much research in the last few years [8,[35][36][37][38]. Moreover, it contains five pre-defined cross-validation splits, making it straightforward to compare our results with that of other research since the data we used for training and testing is the same used in other works. ...
... With the λ = 0 setup, both our original and improved frameworks apply the standard majority voting, as there is no connection between any of the models during training, which leads to each model being trained separately. Therefore, our tables refer to the standard majority voting approach as λ = 0. We also considered several other works that used the same dataset, both traditional [38] and deep learning-based ones [35][36][37]45], and used them as simple baselines and compared their reported results with the performance of our frameworks. For [45], we considered two different methods: one that uses the GLCM and VGG-16 features as described in [45], and an additional one, which follows the same procedure as described in the original paper, but only uses the GLCM matrix as inputs. ...
Article
Full-text available
Brain tumors pose a serious threat in our modern society, with a clear increase in global cases each year. Therefore, developing robust solutions that could automatically and reliably detect brain tumors in their early stages is of utmost importance. In our paper, we revisit the problem of building performant ensembles for clinical usage by maximizing the diversity of the member models during the training procedure. We present an improved, more robust, extended version of our framework and propose solutions that could be integrated into a Computer-Aided Diagnosis system to accurately classify some of the most common types of brain tumors: meningioma, glioma, and pituitary tumors. We show that the new framework based on the histogram loss can be seen as a natural extension of the former approach, as it also calculates the inner products of the latent vectors produced by each member to measure similarity, but at the same time, it also makes it possible to capture more complex patterns. We also present several variants of our framework to incorporate member models with varying dimensional feature vectors and to cope with imbalanced datasets. We evaluate our solutions on a clinically tested dataset of 3,064 T1-weighted contrast-enhanced magnetic resonance images and show that they greatly outperform other state-of-the-art approaches and the base architectures as well, achieving over 92% accuracy, 92% macro and weighted precision, 91% macro and 92% weighted F1\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$F_{1}$$\end{document} score, and over 90% macro and 92% weighted sensitivity.
... Usually, magnetic resonance imaging (MRI) is used to capture the state and structure of the brain and spinal cord to detect and diagnose brain tumors. It is one of the popular medical imaging techniques that employ computer-generated radio waves and magnetic fields to create detailed images of the organs and tissues in the human body [1]. These MR images can be used to easily identify the presence of tumors and their shapes. ...
... Similarly, various types of Deep CNN models were proposed for brain tumor classification. A custom CNN model was designed and implemented to classify multi-grade brain tumors [1]. In contrast, instead of using a single CNN, a hybrid neural network architecture with CNN and DNN was implemented and trained on brain MR images for tumor classification [50]. ...
Article
Full-text available
Accurate medical image classification poses a significant challenge in designing expert computer-aided diagnosis systems. While deep learning approaches have shown remarkable advancements over traditional techniques, addressing inter-class similarity and intra-class dissimilarity across medical imaging modalities remains challenging. This work introduces the advanced gating transformer network (MedTransNet), a deep learning model tailored for precise medical image classification. MedTransNet utilizes channel and multi-gate attention mechanisms, coupled with residual interconnections, to learn category-specific attention representations from diverse medical imaging modalities. Additionally, the use of gradient centralization during training helps in preventing overfitting and improving generalization, which is especially important in medical imaging applications where the availability of labeled data is often limited. Evaluation on benchmark datasets, including APTOS-2019, Figshare, and SARS-CoV-2, demonstrates effectiveness of the proposed MedTransNet across tasks such as diabetic retinopathy severity grading, multi-class brain tumor classification, and COVID-19 detection. Experimental results showcase MedTransNet achieving 85.68% accuracy for retinopathy grading, 98.37% (±0.44\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\pm \,0.44$$\end{document}) for tumor classification, and 99.60% for COVID-19 detection, surpassing recent deep learning models. MedTransNet holds promise for significantly improving medical image classification accuracy.
... In this work, authors improve the performance by using the context information through the spatial relationships between the tumor and its surrounding tissues. On the other hand, Abiwinanda et al. [20] generated seven distinct CNN variations without segmentation for brain tumor classification and achieved the optimal training and testing accuracy of 98.51% and 84.19% on the Fighsare brain MRI dataset, respectively. ...
Article
Full-text available
The recent widespread increase of the Mpox (formerly monkeypox) virus infections in South Asian and African countries has raised concerns among medical professionals regarding the potential emergence of another pandemic in those regions. According to the World Health Organization (WHO) “emergency meeting” on May 20, 2022, there were 82,809 confirmed cases reported in 110 countries. With the number of available test kits surpassing the count of positive/probable cases, there is a pressing need to develop a robust and lightweight classifier model that can alleviate the burden of physical testing kits and expedite the detection process. The existing state-of-the-art primarily focuses on achieving high accuracy in modeling Mpox without considering factors such as modeling suitability, real-time inferencing, and adaptability to resource-constrained CPU-only mobile devices. In this research, we propose a novel lightweight binarized DarkNet53 model, referred to as BinaryDNet53, which is approximately ∼20×\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\sim 20\times $$\end{document} more computationally efficient and ∼2×\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\sim 2\times $$\end{document} more power-efficient than the current state-of-the-art. This model demonstrates smooth detection capabilities when deployed on small hand-held or embedded devices. Firstly, we binarize the weights and biases of the DarkNet53 model to prevent high computational costs and memory usage. Next, our work introduces large-margin feature learning and weighted loss calculation to enhance results, particularly on complex samples. We conduct experiments using the latest MSLD v2.0 dataset, showcasing the superiority of the proposed model over state-of-the-art models based on classification and computational metrics, including Watt power consumption, required memory, and GFLOPS.
... The presented ensemble architecture attained 99.03% accuracy. Abiwinanda et al. (2019) attempted to classify MRI-based brain tumors by developing an optimized CNN architecture. By this network, the authors achieved a detection rate of 84.19%. ...
Article
Full-text available
Introduction Brain cancer is a frequently occurring disease around the globe and mostly developed due to the presence of tumors in/around the brain. Generally, the prevalence and incidence of brain cancer are much lower than that of other cancer types (breast, skin, lung, etc.). However, brain cancers are associated with high mortality rates, especially in adults, due to the false identification of tumor types, and delay in the diagnosis. Therefore, the minimization of false detection of brain tumor types and early diagnosis plays a crucial role in the improvement of patient survival rate. To achieve this, many researchers have recently developed deep learning (DL)-based approaches since they showed a remarkable performance, particularly in the classification task. Methods This article proposes a novel DL architecture named BrainCDNet. This model was made by concatenating the pooling layers and dealing with the overfitting issues by initializing the weights into layers using ‘He Normal’ initialization along with the batch norm and global average pooling (GAP). Initially, we sharpen the input images using a Nimble filter, which results in maintaining the edges and fine details. After that, we employed the suggested BrainCDNet for the extraction of relevant features and classification. In this work, two different forms of magnetic resonance imaging (MRI) databases such as binary (healthy vs. pathological) and multiclass (glioma vs. meningioma vs. pituitary) are utilized to perform all these experiments. Results and discussion Empirical evidence suggests that the presented model attained a significant accuracy on both datasets compared to the state-of-the-art approaches, with 99.45% (binary) and 96.78% (multiclass), respectively. Hence, the proposed model can be used as a decision-supportive tool for radiologists during the diagnosis of brain cancer patients.
... Nyoman and other researchers have explored the use of convolutional neural network (CNNs) for automated tumor classification. They employed a simple CNN architecture consisting of basic layers for convolution, max-pooling, and flattening, followed by a single hidden layer [2]. Despite its simplicity, the CNN achieved impressive results when trained on a dataset of T1 weighted CE-MRI images of glioma, meningioma, and pituitary tumors. ...
Article
Brain tumor classification is a critical task in medical imaging, aiding in timely diagnosis and treatment planning. In this paper, we propose a comprehensive approach utilizing deep learning models for the classification of brain tumors into four categories: glioma, meningioma, pituitary tumors, and no tumors. We employ state-of-the-art convolutional neural network (CNN) architectures including ResNet50, DenseNet201, EfficientNetB3, and InceptionV3 for the classification task. To enhance the performance of the model, we employ resizing and augmentation techniques such as flips and rotation, thereby increasing the diversity of the training dataset. This is particularly crucial due to the limitations posed by small-sized datasets in previous methodologies. Our findings underscore the efficacy of deep learning approaches in brain tumor classification, with EfficientNetB3 emerging as a promising model for accurate diagnosis. Furthermore, our utilization of resizing and augmentation techniques demonstrates their significance in mitigating the challenges associated with limited training data.
Article
Full-text available
INTRODUCTION: Brain tumors have become a major global health concern, characterized by the abnormal growth of brain cells that can negatively affect surrounding tissues. These cells can either be malignant (cancerous) or benign (non-cancerous), with their impact varying based on their location, size and type. OBJECTIVE: Early detection and classification of brain tumors are challenging due to their complex and variable structural makeup. Accurate early diagnosis is crucial to minimize mortality rates. METHOD: To address this challenge, researchers proposed an optimized model based on Convolutional Neural Networks (CNNs) with transfer learning, utilizing architectures like Inception-V3, AlexNet, VGG16, and VGG19. This study evaluates the performance of these adjusted CNN models for brain tumor identification and classification using MRI data. The TCGA-LGG and The TCIA, two well-known open-source datasets, were employed to assess the model's performance. The optimized CNN architecture leveraged pre-trained weights from large image datasets through transfer learning. RESULTS: The refined ResNet50-152 model demonstrated impressive performance metrics: for the non-tumor class, it achieved a precision of 0.98, recall of 0.95, F1 score of 0.93, and accuracy of 0.94; for the tumor class, it achieved a precision of 0.87, recall of 0.92, F1 score of 0.88, and accuracy of 0.96. CONCLUSION: These results indicate that the refined CNN model significantly improves accuracy in classifying brain tumors from MRI scans, showcasing its potential for enhancing early diagnosis and treatment planning.
Article
Full-text available
Brain tumor segmentation in magnetic resonance imaging (MRI) is considered a complex procedure because of the variability of tumor shapes and the complexity of determining the tumor location, size, and texture. Manual tumor segmentation is a time-consuming task highly prone to human error. Hence, this study proposes an automated method that can identify tumor slices and segment the tumor across all image slices in volumetric MRI brain scans. First, a set of algorithms in the pre-processing stage is used to clean and standardize the collected data. A modified gray-level co-occurrence matrix and Analysis of Variance (ANOVA) are employed for feature extraction and feature selection, respectively. A multi-layer perceptron neural network is adopted as a classifier, and a bounding 3D-box-based genetic algorithm is used to identify the location of pathological tissues in the MRI slices. Finally, the 3D active contour without edge is applied to segment the brain tumors in volumetric MRI scans. The experimental dataset consists of 165 patient images collected from the MRI Unit of Al-Kadhimiya Teaching Hospital in Iraq. Results of the tumor segmentation achieved an accuracy of 89% ± 4.7% compared with manual processes.
Article
Full-text available
Content-based image retrieval (CBIR) techniques have currently gained increasing popularity in the medical field because they can use numerous and valuable archived images to support clinical decisions. In this paper, we concentrate on developing a CBIR system for retrieving brain tumors in T1-weighted contrast-enhanced MRI images. Specifically, when the user roughly outlines the tumor region of a query image, brain tumor images in the database of the same pathological type are expected to be returned. We propose a novel feature extraction framework to improve the retrieval performance. The proposed framework consists of three steps. First, we augment the tumor region and use the augmented tumor region as the region of interest to incorporate informative contextual information. Second, the augmented tumor region is split into subregions by an adaptive spatial division method based on intensity orders; within each subregion, we extract raw image patches as local features. Third, we apply the Fisher kernel framework to aggregate the local features of each subregion into a respective single vector representation and concatenate these per-subregion vector representations to obtain an image-level signature. After feature extraction, a closed-form metric learning algorithm is applied to measure the similarity between the query image and database images. Extensive experiments are conducted on a large dataset of 3604 images with three types of brain tumors, namely, meningiomas, gliomas, and pituitary tumors. The mean average precision can reach 94.68%. Experimental results demonstrate the power of the proposed algorithm against some related state-of-the-art methods on the same dataset.
Research
A Convolutional Neural Network (CNN) is a deep neural network architecture inspired by the visual cortex of the human brain, that can learn invariant features from an input matrix. It is used for a number of applications such as image and video recognition, classification and analysis, recommender systems, natural language processing, speech recognition and time series analysis. In general, CNNs consist of alternating convolutional and feature pooling layers followed by fully-connected ones. Besides the ability to learn invariant features, their advantage lies in the reduction of trainable parameters allowing higher efficiency in terms of memory and complexity of processing. In this work, we explain and define all the elements and parameters related to CNNs, including layer design, training, regularization, and optimization. In addition, we present some of the state-of-the-art CNNs and compare their performance.
Article
Among brain tumors, gliomas are the most common and aggressive, leading to a very short life expectancy in their highest grade. Thus, treatment planning is a key stage to improve the quality of life of oncological patients. Magnetic Resonance Imaging (MRI) is a widely used imaging technique to assess these tumors, but the large amount of data produced by MRI prevents manual segmentation in a reasonable time, limiting the use of precise quantitative measurements in the clinical practice. So, automatic and reliable segmentation methods are required; however, the large spatial and structural variability among brain tumors make automatic segmentation a challenging problem. In this paper, we propose an automatic segmentation method based on Convolutional Neural Networks (CNN), exploring small 33 kernels. The use of small kernels allows designing a deeper architecture, besides having a positive effect against overfitting, given the fewer number of weights in the network. We also investigated the use of intensity normalization as a pre-processing step, which though not common in CNN-based segmentation methods, proved together with data augmentation to be very effective for brain tumor segmentation in MRI images. Our proposal was validated in the Brain Tumor Segmentation Challenge 2013 database (BRATS 2013), obtaining simultaneously the first position for the complete, core, and enhancing regions in Dice Similarity Coefficient metric (0:88, 0:83, 0:77) for the Challenge data set. Also, it obtained the overall first position by the online evaluation platform. We also participated in the on-site BRATS 2015 Challenge using the same model, obtaining the second place, with Dice Similarity Coefficient metric of 0:78, 0:65, and 0:75 for the complete, core, and enhancing regions, respectively.
Article
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States in the current year and compiles the most recent data on cancer incidence, mortality, and survival. Incidence data were collected by the National Cancer Institute (Surveillance, Epidemiology, and End Results [SEER] Program), the Centers for Disease Control and Prevention (National Program of Cancer Registries), and the North American Association of Central Cancer Registries. Mortality data were collected by the National Center for Health Statistics. In 2016, 1,685,210 new cancer cases and 595,690 cancer deaths are projected to occur in the United States. Overall cancer incidence trends (13 oldest SEER registries) are stable in women, but declining by 3.1% per year in men (from 2009-2012), much of which is because of recent rapid declines in prostate cancer diagnoses. The cancer death rate has dropped by 23% since 1991, translating to more than 1.7 million deaths averted through 2012. Despite this progress, death rates are increasing for cancers of the liver, pancreas, and uterine corpus, and cancer is now the leading cause of death in 21 states, primarily due to exceptionally large reductions in death from heart disease. Among children and adolescents (aged birth-19 years), brain cancer has surpassed leukemia as the leading cause of cancer death because of the dramatic therapeutic advances against leukemia. Accelerating progress against cancer requires both increased national investment in cancer research and the application of existing cancer control knowledge across all segments of the population. CA Cancer J Clin 2016. © 2016 American Cancer Society.