Genomic Predictors of Drug Sensitivity in Cancer: Integrating Genomic Data for Personalized Medicine in the USA

Аннотация

Despite applying conventional predictive methodologies to obtain genomic insights, predicting drug sensitivity for healthcare organizations in the USA remains a daunting challenge. Cancer is a highly dynamic, adaptive disease tumor cells have repeatedly shown the capability to evolve mechanisms whereby therapeutic interventions can be evaded. Besides, one genomic alteration seldom predicts drug sensitivity. This research project aimed to address the challenges of predicting drug sensitivity by leveraging the GDSC dataset, an extensive resource connecting genomic profiles of cancer cell lines with their sensitivity to a wide range of anti-cancer drugs. This research's key focus was identifying robust genomic markers, including any specific mutations, gene expression patterns, or epigenetic modifications associated with drug sensitivity or resistance. Advanced machine learning and statistical methods were utilized by the predictive models to analyze complex relations that may exist between different genomic alterations and their drug sensitivity. The dataset used for this research project was derived from the Kaggle website. This dataset was compiled by the research project Genomics of Drug Sensitivity in Cancer collaboration between the Sanger Institute in the United Kingdom and the Massachusetts General Hospital Cancer Center in the United States. In their investigation, there was a massive screening of human cancer cell lines with a wide range of anti-cancer drugs. Data collection was performed by large-scale screening of diverse anti-cancer drugs against human cancer cell lines of various types. Cell viability was measured using the Cell-Titer-Glo assay following 72 hours of drug treatment. Several machine learning models were deployed, namely, Random Forest, Linear Regression, and XG-Boost, which exhibited specific strengths. Specific performance metrics used included MSE, RMSE, MAE, and R². As the statistics indicate, among the three models, Random Forest stands out and performs the best on this dataset across all metrics. A smaller value of MAE, MSE, and RMSE signifies that it provided the best forecast for the target variable. It also gave the highest R-squared value. Application of drug sensitivity prediction analysis in cancer can provide an overview of the mechanisms that underlie both tumor response and resistance by investigating the model predictions. The proposed predictive models have the potential to make significant impacts on clinical decision-making in cancer therapy. Predictive models derive informed decisions regarding a patient's risk of recurrence of disease, their response to certain therapies, and their prognosis based on complex clinical and genomic data.

Тип источника: Журналы
Годы охвата с 2020
inLibrary
Google Scholar
Выпуск:
CC BY f
1-21
135

Скачивания

Данные скачивания пока недоступны.
Поделиться
pant, L., Mukaddim, A. A., Rahman, M. K. R., Sayeed, A. A. S., Hossain, M. S. H., Khan, M. T. K., & Ahmed, A. (2024). Genomic Predictors of Drug Sensitivity in Cancer: Integrating Genomic Data for Personalized Medicine in the USA. in Library, 1(3), 1–21. извлечено от https://inlibrary.uz/index.php/archive/article/view/48360
Crossref
Сrossref
Scopus
Scopus

Аннотация

Despite applying conventional predictive methodologies to obtain genomic insights, predicting drug sensitivity for healthcare organizations in the USA remains a daunting challenge. Cancer is a highly dynamic, adaptive disease tumor cells have repeatedly shown the capability to evolve mechanisms whereby therapeutic interventions can be evaded. Besides, one genomic alteration seldom predicts drug sensitivity. This research project aimed to address the challenges of predicting drug sensitivity by leveraging the GDSC dataset, an extensive resource connecting genomic profiles of cancer cell lines with their sensitivity to a wide range of anti-cancer drugs. This research's key focus was identifying robust genomic markers, including any specific mutations, gene expression patterns, or epigenetic modifications associated with drug sensitivity or resistance. Advanced machine learning and statistical methods were utilized by the predictive models to analyze complex relations that may exist between different genomic alterations and their drug sensitivity. The dataset used for this research project was derived from the Kaggle website. This dataset was compiled by the research project Genomics of Drug Sensitivity in Cancer collaboration between the Sanger Institute in the United Kingdom and the Massachusetts General Hospital Cancer Center in the United States. In their investigation, there was a massive screening of human cancer cell lines with a wide range of anti-cancer drugs. Data collection was performed by large-scale screening of diverse anti-cancer drugs against human cancer cell lines of various types. Cell viability was measured using the Cell-Titer-Glo assay following 72 hours of drug treatment. Several machine learning models were deployed, namely, Random Forest, Linear Regression, and XG-Boost, which exhibited specific strengths. Specific performance metrics used included MSE, RMSE, MAE, and R². As the statistics indicate, among the three models, Random Forest stands out and performs the best on this dataset across all metrics. A smaller value of MAE, MSE, and RMSE signifies that it provided the best forecast for the target variable. It also gave the highest R-squared value. Application of drug sensitivity prediction analysis in cancer can provide an overview of the mechanisms that underlie both tumor response and resistance by investigating the model predictions. The proposed predictive models have the potential to make significant impacts on clinical decision-making in cancer therapy. Predictive models derive informed decisions regarding a patient's risk of recurrence of disease, their response to certain therapies, and their prognosis based on complex clinical and genomic data.


background image












Genomic Predictors of Drug Sensitivity in Cancer: Integrating

Genomic Data for Personalized Medicine in the USA

Laxmi pant

1

, Abdullah Al Mukaddim

2

, Md Khalilor Rahman

3

, Abdullah AL Sayeed

4

, Md

Sazzad Hossain

5

, MD Tushar Khan

6

and Adib Ahmed

7

135

MBA in Business Analytics, Gannon University, Erie, PA, USA

2

Master of Science in Business Analytics, Grand Canyon University, Phoenix, AZ, USA

4

Master of Business Administration in Project Management, Central Michigan University, Mt

Pleasant, MI, USA

6

Master of Science in Business Analytics, Trine University, Angola, IN, USA

7

Department of Management Science and Quantitative Methods, Gannon University, Erie, PA,

USA

Corresponding Author:

Laxmi Pant, E-mail: Pant001@gannon.edu

Article Received:01-07-24

Accepted:30-10-24

Published:15-11-24

Licensing Details: Author retains the right of this article. The article is distributed under the
terms of the Creative Commons Attribution-NonCommercial 4.0 License
(http://www.creativecommons.org/licences/by-nc/4.0/),which permits non-commercial use,
reproduction and distribution of the work without further permission provided the original
work is attributed as specified on the Journal open access page

Abstract

Despite applying conventional predictive methodologies to obtain genomic insights,

predicting drug sensitivity for healthcare organizations in the USA remains a daunting challenge.
Cancer is a highly dynamic, adaptive disease tumor cells have repeatedly shown the capability to
evolve mechanisms whereby therapeutic interventions can be evaded. Besides, one genomic
alteration seldom predicts drug sensitivity. This research project aimed to address the challenges
of predicting drug sensitivity by leveraging the GDSC dataset, an extensive resource connecting
genomic profiles of cancer cell lines with their sensitivity to a wide range of anti-cancer drugs.


background image

This research's key focus was identifying robust genomic markers, including any specific
mutations, gene expression patterns, or epigenetic modifications associated with drug sensitivity
or resistance. Advanced machine learning and statistical methods were utilized by the predictive
models to analyze complex relations that may exist between different genomic alterations and their
drug sensitivity. The dataset used for this research project was derived from the Kaggle website.
This dataset was compiled by the research project Genomics of Drug Sensitivity in Cancer
collaboration between the Sanger Institute in the United Kingdom and the Massachusetts General
Hospital Cancer Center in the United States. In their investigation, there was a massive screening
of human cancer cell lines with a wide range of anti-cancer drugs. Data collection was performed
by large-scale screening of diverse anti-cancer drugs against human cancer cell lines of various
types. Cell viability was measured using the Cell-Titer-Glo assay following 72 hours of drug
treatment. Several machine learning models were deployed, namely, Random Forest, Linear
Regression, and XG-Boost, which exhibited specific strengths. Specific performance metrics used
included MSE, RMSE, MAE, and R². As the statistics indicate, among the three models, Random
Forest stands out and performs the best on this dataset across all metrics. A smaller value of MAE,
MSE, and RMSE signifies that it provided the best forecast for the target variable. It also gave the
highest R-squared value. Application of drug sensitivity prediction analysis in cancer can provide
an overview of the mechanisms that underlie both tumor response and resistance by investigating
the model predictions. The proposed predictive models have the potential to make significant
impacts on clinical decision-making in cancer therapy. Predictive models derive informed
decisions regarding a patient's risk of recurrence of disease, their response to certain therapies, and
their prognosis based on complex clinical and genomic data.

Keywords:

Genomic Predictors; Drug Sensitivity; Genomic Markers; Personalized medicine;

Machine Learning; Random Forest Algorithm

INTRODUCTION

Background and Motivation

According to Bortty (2024), the protocols of cancer treatment have witnessed a

transformative shift in the USA with the emergence of personalized medicine, which curates’
therapeutic strategies to the individual characteristics of each patient. These procedures are of
paramount importance in oncology because of the very essence of molecular heterogeneity among
an individual's cancers, which could have a great influence on the modality of treatment applied.
Al Amin (2024), contended that Personalized medicine in oncology does take cognizance that two
patients diagnosed with the same histological kind of cancer could react very differently to
identical therapies. These differences are very often underlined by genomic differences, which
include mutations, gene expression level, copy number alteration, and other molecular changes
that underlie the behavior of a tumor and its responsiveness to drugs. Chakravarty & Solit (2024),
postulated that genomic data serves as a cornerstone for understanding drug sensitivity and
resistance in cancer. By highlighting the molecular oncogenic drivers of cancer, and their


background image

interaction with specific therapeutics, genomic data enables the oncologist to select the most active
treatment options, thus minimizing the unnecessary toxicity and improving survival.

Notwithstanding, Bhomik et al. (2024), argued that despite its promise, the application of

genomic insights to predict drug sensitivity in the USA remains a daunting challenge. Cancer is a
highly dynamic, adaptive disease tumor cells have repeatedly shown the capability to evolve
mechanisms whereby therapeutic interventions can be evaded. Besides, Hider et al. (2024), added
that one genomic alteration seldom predicts drug sensitivity; it is a product of complex interactions
in the molecular network of a given tumor. Added layers of complexity come from variability
between different patients, environmental factors, and influences from the tumor
microenvironment that together make response to treatment unpredictable. These complexities
have been enabled to be probed by the availability of high-throughput genomic datasets, including
The Cancer Genome Atlas and the Genomics of Drug Sensitivity in Cancer. Dutta et al. (2024),
asserted that the integration of such large datasets into actionable clinical insights remains a
significant challenge due to various challenges in data standardization, computation modeling, and
interpretation of a biological system. It is essential to bridge these gaps if the full potential of
genomic data is to be realized in informing personalized oncology.

Objectives

This research project aims to address the challenges of predicting drug sensitivity by

leveraging the GDSC dataset, an extensive resource connecting genomic profiles of cancer cell
lines with their sensitivity to a wide range of anti-cancer drugs. The key focus of this research will
be the identification of robust genomic markers, including any specific mutations, gene expression
patterns, or epigenetic modifications that are associated with drug sensitivity or resistance.
Detection of such markers will therefore elucidate the molecular mechanisms of differential
responses to drugs, hence enabling more specific therapeutic intervention. In addition, this baseline
study is done in a manner that predictive models can be developed that combine genomic data to
guide treatment plans for individuals specifically. Advanced machine learning and statistical
methods will be utilized by the predictive models to analyze complex relations that may exist
between different genomic alterations and their drug sensitivity. By integrating multi-omics data,
namely, genomics, transcriptomics, and epigenomics, the models capture complex biology to offer
better predictiveness. This effort is targeted at its translation into clinical practice to make sure that
treatment decisions by oncologists in the USA can be reached and optimized in all outcomes for
the patients. This study contributes to the broad vision of precision oncology: to deliver the right
drug, at the right dose, to the right patient, at the right time.

LITERATURE REVIEW

Overview of Personalized Medicine in Oncology

Islam et al. (2024), posited that the new paradigms of oncology treatment involve

personalized medicine, emphasizing the need to tailor therapeutic intervention toward a unique
genetic and molecular profile singular to the individual. Nasiruddin et al. (2024), reported that


background image

Traditionally, cancer treatments have been very uniform, largely depending on broad-spectrum
chemotherapeutic drugs and radiation, which in all too many instances provide significant toxicity
yet highly variable efficacy. In contrast, however, personal medicine would take advantage of
recent advances in genomics, proteomics, and bioinformatics to classify patients with specific
biological features of their tumors. This precision-driven approach has redefined cancer care by
enabling the development of targeted therapies and immunotherapies, along with companion
diagnostics that optimize treatment efficacy while minimizing adverse effects (Prabhod 2022).

Rahman et al. (2024), posited that personalized medicine plays a significant role in tackling

the inherent heterogeneity of cancer. Each tumor possesses a unique set of molecular alterations,
including point mutations in key oncogenes and tumor suppressors, epigenetic changes, and
variations in gene expression. These alterations not only are responsible for tumorigenesis but also
modulate how a given tumor will respond to specific treatments. It is for personalized medicine to
decipher this complexity by evolving from the "one-size-fits-all" approach into a more
discriminating strategy wherein therapeutic choices are guided by the molecular profiles of the
tumor (Quazi, 2022). This assures very deep impacts on patient outcomes because it identifies in
advance those patients who will benefit the most and avoids nonspecific prescriptions that lead to
benefits in survival.

Chawla et al. (2022), reported that the modern landscape of personalized cancer treatment

has now drastically expanded, with multiple targeted therapies and immunotherapies approved by
the FDA. Examples include trastuzumab for HER2-positive breast cancer, imatinib for BCR-ABL-
positive chronic myeloid leukemia, and pembrolizumab for cancers with high microsatellite
instability. De Jong et al., (2021), asserted that clinical trials increasingly use genomic profiling to
stratify patients and evaluate the effectiveness of targeted agents within specific molecular
backgrounds. Despite these advances, significant challenges remain to fully individualize medical
treatment in the clinic, particularly in identifying robust biomarkers of drug sensitivity and
resistance. In overcoming these, the full potential of personalized oncology can be fully realized,
and access to precision treatments equitably distributed across diverse populations.

Genomic Predictors and Drug Sensitivity

The genomic alteration and drug sensitivity relationship has been a point of high focus in

cancer research, with normally many attempts to pinpoint biomarkers that predict therapeutic
responses. Including somatic mutations, copy number variations, gene fusions, and epigenetic
modifications, genomic markers are pivots in determining how a tumor interacts with specific
drugs (Li et al. 2021). For instance, mutations in EGFR predict sensitivity to EGFR tyrosine kinase
inhibitors in non-small cell lung cancer and predict resistance to anti-EGFR therapies due to KRAS
mutations in colorectal cancer (Lewis &Kemp, 2021). Likewise, mutations in BRCA1/2 are
predictive biomarkers of the action of PARP inhibitors in ovarian and breast cancers. These
findings emphasize the importance of incorporating genomic insights into clinical decisions to
increase the precision of cancer therapy.


background image

Hou et al.(2024), argued that one of the major resources to date, for elucidation of the

molecular rationale of drug sensitivity, has been provided by the dataset on the genomics of drug
sensitivity in cancer. The GDSC project systematically profiles hundreds of cancer cell lines,
linking their genomic features to sensitivity or resistance against a wide array of anti-cancer drugs.
Indeed, this dataset embodies information from somatic mutations, expression levels of genes,
copy number alterations, and various metrics of drug response; thus, it is a rich scientific resource
for searching biomarkers and therapeutic targets (Quazi, 2022). A large number of studies on the
understanding of the mechanisms of drug sensitivity and resistance could be enabled with the
GDSC dataset through large-scale genomic and pharmacological data.

Thirunavukarasu et al. (2022), articulated that one of the major applications of the GDSC

dataset is in finding novel genomic markers predictive of drug responses across diverse cancers.
The studies using this data from GDSC have pointed out, for example, that the mutations in genes
such as TP53, PIK3CA, and PTEN modulate the sensitivity to concrete therapies. The dataset has
proved instrumental in unraveling the relationships between patterns of gene expression and the
efficacy of particular drugs, and also in validation of the therapeutic potential of newer drugs.
Continuing to drive innovation into personalized cancer therapy, the GDSC project lays out an
integrated framework of genomic and pharmacological data (Li et al. 2021). Translating such
insights into clinical practice, however, requires the use of robust computational approaches
amenable to the analysis of complex, multi-dimensional data generated by high-throughput
profiling technologies.

Machine Learning in Genomic Data Analysis

Al amin et al. (2024), affirmed Machine learning has indisputably revolutionized the

analysis of genomic data, enabling analysis robustly for underlying patterns and relations that
could not be easily captured by traditional statistical methods. The algorithms of machine learning
are good at handling high-dimensional, multi-modal datasets, thus becoming particularly suitable
for handling large data emerging from projects like GDSC. These algorithms can spot complex,
nonlinear relationships between genomic features and drug sensitivity, enabling biomarker and
predictive model identification to inform personalized treatment strategies (Li et al. 2021).

Bhowmik et al. (2024), found that some of the most applied machine-learning approaches

incorporate random forests, XG-Boost, support vector machines, neural networks, and ensemble
learning in cancer genomics for the prediction of drug responses. These algorithms range from
deep learning models that integrate multi-layered input from genomic, transcriptomic, and
epigenomic data to others. Indeed, using such approaches, very high accuracy has been attained in
the prediction of sensitivity to particular drugs. Bortty et al. (2024), argued that other approaches
have used unsupervised learning techniques such as clustering and dimensionality reduction to
classify tumors into molecular subtypes with differential therapeutic vulnerabilities. This therefore
shows that machine learning is indeed capable of deriving actionable insights from complex
datasets and, therefore, driving innovation in precision oncology.


background image

Dutta et al. (2024), pointed out that previous machine learning research on drug sensitivity

prediction gives promising results. For example, a study presented the application of gradient-
boosting algorithms on GDSC data toward the identification of genomic features associated with
sensitivity to over 200 drugs and uncovered novel biomarkers and potential therapeutic targets.
Another recent study combined GDSC data with patient-derived tumor profiles; deep learning was
used to predict clinical response to targeted therapies. These results also indicate the role of
machine learning in bridging the gap existing between preclinical datasets and clinical
applications, hence opening new avenues for more valid and personalized treatments of cancers.
These successes notwithstanding, there are yet some challenges in the implementation of machine
learning models for genomic data analysis (Hider et al., 2024). One major obstacle lies in the fact
that high-quality annotated datasets, which capture the full spectrum of tumor heterogeneity and
drug responses, are still lacking. Moreover, there is a big challenge in the interpretability of
machine learning models, since many algorithms may obscure the actual biologically grounded
mechanism for their predictions. These are challenges that require the development of models
transparently and interpretably, predictive performance is balanced against the plausibility of the
underlying biology. These outcomes further requires synergy between computational scientists,
biologists, and clinicians to ensure that insights driven by machine learning go through clinically
relevant applications (Rahman et al., 2024)

DATA COLLECTION AND PREPROCESSING


Data Sources

The dataset used for this research project was derived from the Kaggle website. This dataset

was compiled by the research project Genomics of Drug Sensitivity in Cancer collaboration
between the Sanger Institute in the United Kingdom and the Massachusetts General Hospital
Cancer Center in the United States (Alipour, 2024). In their investigation, there was a massive
screening of human cancer cell lines with a wide range of anti-cancer drugs. Data collection was
performed by large-scale screening of diverse anti-cancer drugs against human cancer cell lines of
various types. Cell viability was measured using the Cell-Titer-Glo assay following 72 hours of
drug treatment. The datasets are accessible and can be downloaded from the GDSC website itself:
GDSC Database (Alipour, 2024). The Genomics of Drug Sensitivity in Cancer dataset is a very
useful resource in therapeutic biomarker discovery within cancer research. It combined
pharmacological profiles of tumor cell lines' drug response with matched genomic data and
enabled the study of relationships between mutation and copy number variation events and
sensitivity to drugs. The main task related to this dataset was to predict the drug sensitivity-e.g.,
IC50 of the cancer cell line based on its genomic features. That included a regression task to predict
the exact IC50 values or a classification task to predict whether the cell lines are sensitive or
resistant to particular drugs (Alipour 2024). These data can similarly be used to find genomic
markers predictive of drug response. The dataset contained the following columns:


background image

Column

Description

COSMIC_ID:

Unique identifier for each cell line.

CELL_LINE_NAME:

Name of the cell line.

TCGA_DESC:

Description of the TCGA label for cancer types.

DRUG_ID:

Unique identifier for each drug.

DRUG_NAME:

Name of the drug.

LN_IC50:

The logarithm of the half-maximal inhibitory
concentration (target column).

AUC:

The area Under the Curve represents the drug's
efficacy.

Z_SCORE:

Standardized value for the drug's response.

GDSC Tissue descriptors:

Tissue type descriptors for cancer.

Cancer Type (matching TCGA label):

Specific cancer type for the cell line.

Microsatellite instability Status (MSI):

MSI status for cancer cells.

Screen Medium:

Medium is used for drug screening.

Growth Properties:

Properties describing cell growth.

CNA:

Copy number alterations for the cell lines.

Gene Expression:

Gene expression data for cell lines.

Methylation:

DNA methylation data for cell lines.

TARGET:

The putative molecular target for the drug.

TARGET_PATHWAY:

The biological pathway related to the target.

Table 1: Showcases the Columns Contained in the Dataset

Data Preprocessing


Data pre-processing-first checked two datasets, namely GDSC and Compounds Annotation

for missing values. The computed code went through each column in the GDSC dataset, counting
the number of missing values and printing them out. Similarly, it also performed that for the
Compounds Annotation dataset. This step was very important because missing data has a serious
effect on the quality and reliability of subsequent analysis and modeling (Pro-AI-,2024).
Subsequently, imputation techniques such as mean or median imputation, mode imputation, and
more sophisticated techniques like regression or machine learning-based imputation were applied,
depending on the nature of the missing data and the particular goals of the analysis (Pro-AI-, 2024).
The early identification of missing values during the preprocessing stage facilitated informed
decisions on how to handle these values to ensure data integrity and resultant accuracy.






background image

Exploratory Data Analysis

Figure 1: Displays the Distribution of Key Variables, LN IC50, AUC & Z-Score

The graph above showcases the distribution of three key variables: LN IC50, AUC, and Z

SCORE. The distribution of LN IC50 is around normal but has a slight positive skew, indicating
that most of the IC50 values were concentrated while few were higher in the narrow range. AUC
distribution is highly right-skewed, meaning the greatest number of values fall closer to the lower
bound of the scale, while only a few cases have extremely high AUC values. The distribution of Z
SCORE is much like that of a normal distribution in that it is symmetric around zero and captures
an effect within the tails on both sides, showing that values circle the mean. These distributions
give an idea of the range and variability of such variables in this set of data.

Figure 2: Displays the Distribution of Log-Transformed AUC

This graph represents the distribution of log-transformed AUC values. The distribution is

right-skewed, extending with a long tail to the right toward higher values of AUC; it means that
the greater part of the observations have lower AUC scores, while only a minor share has much
higher AUC values. The peak of such a distribution would mean that a lot of observations are
grouped around a particular range of AUC values. Log transformation probably was used to
normalize the distribution and make the distribution more normal for statistical analysis. Even
then, after such a log transformation, right-skewness persists. It indicates that the actual data may
contain some degree of non-normality.


background image

Figure 3: Exhibits the Target Distribution

The bar chart above represents the distribution of target values in a dataset. Different target

categories are on the x-axis, and on the y-axis is the count of occurrences of each category. It
shows the imbalanced nature of the distribution. There are a few categories that form the
dominating portion of this dataset. For the top few categories, their counts are high compared to
the majority of the other categories. This may indicate a potential problem in the construction of
the predictive model, as the model can get biased towards the majority classes and will not predict
the minority classes correctly. This will need to be balanced by either oversampling, under-
sampling, or class weighting to make sure the model is fair and robust.

Figure 4: Portrays the Distribution of Log-Transformed AUC

This graph represents the distribution of log-transformed AUC values. The distribution

appears right-skewed; it contains a prolonged tail at higher values of AUC. That would suggest
that a greater proportion of the observations have lower AUC scores, while the rest have much
higher AUC values. The peak shows that most of the observations cluster around a specific range
of values of AUC. This log transformation likely served to normalize the distribution, ultimately
making the data suitable for most statistical analyses. Even then, the right-skewed nature of this


background image

distribution is still present after the transformation, suggesting that there may have been some
degree of non-normality in the raw data.

Figure 5: Depicts the Distribution of the Three Key Variables

The graphs above display the distribution of three important variables: LN IC50, AUC, and

Z SCORE. The distribution of LN IC50 is approximately normal with minor positive skew; this
indicates that most of the IC50 values fall under a particular range of values. From this histogram,
the distribution of AUC is highly right-skewed, indicating that there are considerable values
huddled in the lesser scale of the variable with a few instances recording high AUC values. The
distribution of Z SCORE is approximately normal since it has a hump around zero and symmetric
tails, which would suggest that the values are strewn around the mean with variations on both
sides. These distributions give an insight into the range and variability of the dataset for these
variables.

METHODOLOGY

Feature Engineering and Selection

Feature engineering is the process of generating raw genomic data into a format that the

machine learning model will need so that the model fully understands the underlying pattern linked
with drug sensitivity. This protocol is especially important in a high-dimensional genomic dataset
such as the Genomics of Drug Sensitivity in Cancer that contains exhaustive data regarding genetic
mutations, copy number variations, and epigenetic profiles of gene expressions. Suitable
preprocessing, feature extraction, and refinement have been performed to enhance the predictive
power of machine learning models. In that respect feature extraction methods summarized
complicated genomic data into interpretable metrics. Features were pre-aggregated across, for
instance, biological pathways or functional gene groups rather than individual gene expression
values. Dimensionality was reduced by a variety of techniques, such as PCA or autoencoders, that
retain the most informative patterns present in the data. Specifically, PCA defined the directions
within which most of the variances in data are explained. Such methods, therefore, enabled the
turning of thousands of variables into several components in an understandable manner with no
significant loss of information. By contrast, autoencoders represented neural network methods that


background image

learn compressed representations of data through unsupervised learning and are therefore befitting
for the capture of nonlinear relationships between genomic features.

Model Selection Justification

The choice of a suitable and strategic machine learning model is very pivotal in making an

accurate prediction of drug sensitivity because the nature of genomic data requires algorithms that
can deal with high-dimensional, complex, and often noisy inputs. Several machine learning models
were deployed, namely, Random Forest, Linear Regression, and XG-Boost, which exhibited
specific strengths. Firstly, logistic regression served as the standard baseline method for binary
classification, such as the induction of either sensitivity or resistance against a particular drug. This
algorithm was selected because it is simple, interpretable, and effective when the relationship
between the features and outcomes is linear. However, logistic regressions can be fussy with high-
dimensional data when the number of features exceeds the number of samples. On the other hand,
tree-based methods such as Random Forest and XG-Boost-classifiers are befitting for large and
noisy data sets. Random Forest builds an ensemble of decision trees and then averages their
predictions to reduce overfitting and improve generalization. XG-Boost extends it with tree
construction optimization by gradient boosting, which makes the training faster while yielding
higher predictive accuracy.

Training and Testing Framework

The training and testing framework play an indispensable role in affirming that machine

learning algorithms generalize well to undetected data, a paramount requirement for robust drug
sensitivity prediction. Essentially, the dataset was split based on the task at hand into training
subsets, validation, and testing to achieve this. The training set was employed to train the model,
while the validation set was utilized in the tuning of model hyper-parameters and preventing
overfitting of the model. The final evaluation is done on the test set, which remains unseen during
the training and validation phases. Common strategies for splitting include random splitting and
stratified splitting, with the latter ensuring that the class distribution in the subsets reflects the
original dataset.

Cross-validation techniques were implemented in the study as integral procedures to model

evaluation, particularly when datasets are small or imbalanced. K-fold cross-validation first splits
the data into k-folds of equal size. The model was trained on k-1 folds, reserving the remaining
fold for the validation set. Repeat the process k times: each fold acts once as a validation set. The
results of the process were averaged to yield a reliable estimate of the performance of the model.
Another approach was the leave-one-out cross-validation, which relies on all but one sample for
training and uses the excluded sample for validation, repeating for all samples in the dataset.
Although computationally more expensive, LOOCV gives one of the better estimates of the
performance of a model.



background image

Hyperparameter Tuning


Hyperparameter tuning is an imperative step in optimizing machine learning algorithms

for genomic data analysis, as the choice of hyperparameters significantly impacts performance.
This is also because the choice of different hyperparameters will notably change the performance.
The hyperparameters are parameters that are fixed before training and determine how the learning
algorithm will behave; examples include the neural net learning rate, the number of trees in a
Random Forest, or the penalty parameter in the case of SVMs.

The best hyperparameters were chosen by exploring the space of hyperparameters

systematically with techniques such as grid search and random search. The Grid Search
systematically attempted all the combinations of hyperparameters within a prespecified range to
ensure that the entire space has been spanned. For example, it could vary the number of trees, the
maximum depth of trees, or the minimum number of samples at each leaf for the optimal selection
in a Random Forest. While usually powerful, grid search is prohibitively computationally
intensive, especially for models with a large number of hyperparameters.

A more efficient alternative is random search, which samples hyperparameter

combinations randomly. Essentially, it has been found that random search outperforms grid search
when only a small number of evaluations can be performed since one covers a wider range of
configurations. Even more advanced is Bayesian optimization, which uses probabilistic models to
predict the performance of hyperparameter configurations and iteratively updates the search with
knowledge from previous measurements.


Performance Metrics


Robust and comprehensive metrics that correctly reflect the model's capability to

generalize to unseen data are of prime importance when the performance of predictive models in
drug sensitivity analysis is to be evaluated. Different metrics employed therefore usually depend
on whether the nature of the prediction task is regression or classification. In regression tasks,
when the model is supposed to predict continuous scores regarding drug sensitivity, examples of
metrics used included MSE, RMSE, MAE, and R². MSE is a measure of the average of the squared
difference between predicted and actual values. Since larger errors are emphasized more by this
squaring operation, it is useful for penalizing models that produce significant outliers. As the
square root of MSE, the RMSE is an interpretable measure that will have the same unit as your
original data, making it easier to contextualize model accuracy. In contrast, MAE computes the
average of the absolute differences between the predictions and actual values; hence, it provides a
straightforward and less sensitive measure of error, especially when outliers are present. R-squared
gives the proportion of variance in a target variable explained by the model. Thus, it is an intuitive
measure of the explanatory power of the model. A high value of R-squared means that most of the
data variability is captured by the model, while low values result from poor performances or failure
in the capture of important relationships.


background image

RESULTS

Descriptive Analysis

Model

MAE

MSE

RMSE

R-Squared[R

2

]

Linear Regression

1.09

2.50

1.58

0.69

Random Forest Regressors

0.16

0.08

0.29

0.99

Gradient Boosting Regressor

0.26

0.13

0.36

0.98

Table 2: Visualizes the Performance Metric of the three Models

The above table presents the evaluated performance metrics of three regression models:

Linear Regression, Random Forest, and Gradient Boosting. The statistics replayed against each
model involve Mean Absolute Error-Mean Squared Error (MAE); Mean Squared Error (MSE);
Root Mean Squared Error (RMSE); and R-squared (R

2

). As the statistics indicate, among the three

models, Random Forest stands out and performs the best on this dataset across all metrics. A
smaller value of MAE [0.16], MSE [0.08], and RMSE [0.29] signifies that it provided the best
forecast for the target variable. It also gave the highest R-squared value [0.99], meaning much
more of the variation in data could be explained by the algorithm. It was observed that Linear
Regression gave the poorest result, with high values for MAE [1.09], MSE [2.50], and RMSE
[1.58]. Gradient Boosting reached the middle level, showing an average increase compared to
Linear Regression but falling behind Random Forest ultimately. Overall, the table shows the
superiority of the Random Forest model on this regression task.

Model Performance
a) Random Forest

Table 3: Depicts the Random Forest Modelling

# Step 4: Model Training

pipeline

.

fit(X_train, y_train)

# Step 5: Predictions

y_pred

=

pipeline

.

predict(X_test)

# Step 6: Model Evaluation

def

evaluate_model(predictions, true_values):

mse

=

mean_squared_error(true_values, predictions)

rmse

=

np

.

sqrt(mse)

mae

=

mean_absolute_error(true_values, predictions)

r2

=

r2_score(true_values, predictions)

return

mse, rmse, mae, r2

# Evaluate RandomForestRegressor

rf_mse, rf_rmse, rf_mae, rf_r2

=

evaluate_model(y_pred, y_test)

# Print evaluation results

print("Random Forest Regressor Evaluation:")
print(f"MSE: {rf_mse}, RMSE: {rf_rmse}, MAE: {rf_mae}, R2: {rf_r2}")


background image

The Python code snippet above implemented the machine learning pipeline for regression

tasks. First, it defined the pipeline linking together some preprocessing already enclosed in the
preprocessor variable with a Random Forest Regressor model. Explicitly, the pipeline made sure
that the preprocessing part occurred on the data before the actual training of the model. Then, it
trained the model on preprocessed training data and made predictions on test data. Subsequently,
the function evaluate_model computed four of the most common regression metrics: MSE, root
mean squared error, mean absolute error, and R-squared for assessing a model's performance on
test data. Eventually. It printed the calculated metrics to show the model evaluation concerning the
accuracy and predictive power.

Output:

Table 4: Presents the Random Forest Evaluation Results

Random Forest Regressor Evaluation:

MSE: 0.08252052733612429,
RMSE: 0.28726386360996453,
MAE: 0.1586659554659184,
R2: 0.9897738225597844


The above is the evaluation results for the Random Forest Regressor model. The metrics

in the above figure include Mean Squared Error (MSE), Root Mean Squared Error (RMSE), Mean
Absolute Error (MAE), and R-squared (R

2

). From the R-squared value of 0.9897, this model

explains most of the variance within the data. The low values for MSE and RMSE indicate that
the model simulations are quite close to the actual values. Lower values of such scores require
smaller margins of error in predictions. The MAE, representing the average absolute difference of
the predictions from the actual value, is also quite low. In essence, all these metrics put together
suggest that the Random Forest Regressor performed well on this given dataset.

b) Linear Regression


This snippet below is a Python code that builds an efficient pipeline to pre-process and

model a linear regression. The pipeline-named pipeline_lr-consisted of two steps: a preprocessor,
'preprocessor', which included some form of data preprocessing like scaling, encoding, or some
sort of feature transformation, and the actual linear regression model, 'model', LinearRegression().
It fitted the pipeline to the training dataset, X_train, and y_train, using the fit method and making
predictions on the test set, X_test with the predict method. The resulting predictions, lr_pred are
passed to an evaluation function, evaluate_model which returns the following four performance
metrics: Mean Squared Error-MSE, Root Mean Squared Error-RMSE, Mean Absolute Error-
MAE, and R-squared -R². The last section prints, in tabulated format, the results of these tests.
This code is an example of how pipelines can simplify model training and testing with guaranteed
consistency in preprocessing, making it ideal for scalable and reproducible machine-learning
workflows.


background image

Table 5: Portrays the Linear Regression Modelling.

# Linear Regression Model (in the same pipeline format)

pipeline_lr

=

Pipeline(steps

=

[

('preprocessor', preprocessor),
('model', LinearRegression())
])

pipeline_lr

.

fit(X_train, y_train)

lr_pred

=

pipeline_lr

.

predict(X_test)

lr_mse, lr_rmse, lr_mae, lr_r2

=

evaluate_model(lr_pred, y_test)


print("\nLinear Regression Evaluation:")
print(f"MSE: {lr_mse}, RMSE: {lr_rmse}, MAE: {lr_mae}, R2: {lr_r2}")

Output:

Table 6: Showcases the Linear Regression Evaluation Results

Linear Regression Evaluation:

MSE: 2.498517638627196,
RMSE: 1.5806699967504907,
MAE: 1.0896249837375305,
R2: 0.690376618583177

Above are the evaluation results for a Linear Regression model. Some of the metrics shown

in this output are Mean Squared Error (MSE), Root Mean Squared Error (RMSE), Mean Absolute
Error (MAE), and R-squared (R2). Therefore, from this, the R-squared value is 0.69, and so the
model explains about 69% of the variance in data, which is fairly reasonable. Whereas MSE and
RMSE are variance-like measures of the size of prediction errors, with smaller values indicative
of more accurate models, MAE is like an average absolute difference between predictions and
actual values. The model has a moderate R-square value; hence, further analysis and probably
refinement of the model or inclusion of other variables might well be required to have better
predictive performance.

c) XG-Boost

The code snippet below illustrates the implemented XG-Boost regression model using a

machine-learning pipeline. Steps included in the pipeline are named pipeline_xgb, which mainly
comprises a preprocessor ('preprocessor') for scaling, encoding, or imputing data, and the model
itself, XGB-Regressor. In the model, n_estimators=100 means that 100 decision trees are used in
Boosting; learning_rate=0.1, which controls the contribution amount of each tree during
prediction. Random_state = 42 ensures reproducibility by controlling the randomness involved in
training the model. It trains the pipeline on X_train and y_train with the fit method and makes
predictions on the test set, X_test, using the predict method. This prediction, xgb_pred, is then fed
into an external function, evaluate_model, which calculates several key regression performance
metrics: Mean Squared Error (MSE), Root Mean Squared Error (RMSE), Mean Absolute Error


background image

(MAE), R-squared (R²). At the end, the metrics are printed, which would give an idea about the
model's accuracy and quality of prediction. The above code illustrates the efficiency of pipelines
in embedding the preprocessing with advanced models like XGBoost suitable for complex large-
scale datasets.

Table 7: Portrays the XGBoost Modelling

# XGBoost Model

pipeline_xgb

=

Pipeline(steps

=

[

('preprocessor', preprocessor),
('model', XGBRegressor(n_estimators

=

100, learning_rate

=

0.1,

random_state

=

42))

])

pipeline_xgb

.

fit(X_train, y_train)

xgb_pred

=

pipeline_xgb

.

predict(X_test)

xgb_mse, xgb_rmse, xgb_mae, xgb_r2

=

evaluate_model(xgb_pred, y_test)


print("\nXGBoost Evaluation:")
print(f"MSE: {xgb_mse}, RMSE: {xgb_rmse}, MAE: {xgb_mae}, R2: {xgb_r2}")


Output:

Table 8: Highlights the XG-Boost Evaluation

XGBoost Evaluation:

MSE: 0.1314089752598891,
RMSE: 0.362503758959668,
MAE: 0.25991761196150165,
R2: 0.9837154276441923

The above evaluation results concern the XG-Boost model. The metrics applied are

respectively: Mean Squared Error, Root Mean Squared Error, Mean Absolute Error, and R-
squared. It can be understood that with the high R-squared value of 0.9837, much of the variance
in the data is explained by this model. The small MSE and RMSE indicate that the model's
predicted values are somewhat close to the actual values. Additionally, the MAE of the difference
between the predicted and the actual value is also relatively small. Overall, these metrics together
show that the XG-Boost model performs quite well at making predictions on the provided dataset.


Model Comparison


The following Python code first creates a data frame in which the performance metrics of

three regression models, Random Forest, Linear Regression, and XG-Boost, are kept for
comparison: The data frame consists of the following columns - column names for Model, MSE,
RMSE, MAE, and R

2

. In that respect, the code proceeds to a bar plot, with comparisons of models


background image

in selected performance metrics visual. It plots a bar chart that enables quick and intuitive
comparison of the model performances with the intent of selecting the best for the particular
regression task.

Table 9: Showcases the Plotting of Model Comparison Code Snippet

# Create a DataFrame to store model performance

model_comparison

=

pd

.

DataFrame({

'Model': ['Random Forest', 'Linear Regression', 'XGBoost'],
'MSE': [rf_mse, lr_mse, xgb_mse],
'RMSE': [rf_rmse, lr_rmse, xgb_rmse],
'MAE': [rf_mae, lr_mae, xgb_mae],
'R2': [rf_r2, lr_r2, xgb_r2]
})

# Plotting Model Comparison

model_comparison

.

set_index('Model')

.

plot(kind

=

'bar', figsize

=

(10, 6))

plt

.

title("Model Comparison")

plt

.

ylabel("Score")

plt

.

show()


Output:

Figure 6: Visualizes Model Comparison Histograms

The bar chart above compares the performance of three regression models: Random Forest,

Linear Regression, and XG-Boost, against four evaluating metrics, which are Mean Squared Error,
Root Mean Squared Error, Mean Absolute Error, and R-squared. The plot above discloses that


background image

Random Forest consistently outperforms the other two algorithms across all metrics. It has the
lowest MSE, RMSE, and MAE values, indicating better accuracy in predicting the target variable.
Moreover, it has the highest value of R-squared, which confirms that there is a greater proportion
of explained variance within the data. The worst performance is recorded for Linear Regression
with the highest MSE, RMSE, and MAE values. Gradient Boosting sits between the two; from the
Linear Regression, it improves significantly but remains far behind Random Forest. Overall, this
chart speaks to the superior performance of the Random Forest model on the given regression task.


Predictive Insights

Application of drug sensitivity prediction analysis in cancer can provide an overview of

the mechanisms that underlie both tumor response and resistance by investigating the model
predictions. By investigating the machine learning model predictions, potential drug targets and
biomarkers can be found that predict patient response to particular therapies. It might predict, say,
that for subpopulations of patients with certain genomic profiles, some drugs will work wonders.
A treatment decision can lean on such information and may, therefore, improve patient outcomes.

Additionally, the investigation into the model predictions may reveal novel paired drug

combinations much more potent than single-agent therapies. The identification of synergistic drug
pairs enables the researchers to further develop more specific and personalized treatment regimens.
Also, by considering several model predictions trained on different datasets, the generalizability
of the findings can be tested and potential biases of their data detected.

DISCUSSION

Clinical Implications

Predictive models have the potential to make significant impacts on clinical decision-

making in cancer therapy. Predictive models derive informed decisions regarding a patient's risk
of recurrence of disease, their response to certain therapies, and their prognosis based on complex
clinical and genomic data. This enables the clinician to make better choices for treatment, thus
improving the outcomes for the patients.

Applications of machine learning models in clinical settings enable the extraction of a more

personalized medicine. In such ways, the clinician is able to develop a treatment plan that caters
to the best therapeutic efficacy while at the same time minimizing any potential adverse effects on
the given subject. Based on this premise, for example, high-risk patients would benefit from
aggressive therapy, while low-risk patients would be spared their disposal, among other
unnecessary interventions.

Challenges and Limitations

The application of predictive models into clinical practice opens up several ethical issues.

There is a great need for proper data privacy along with security for the protection of sensitive
genomic data to maintain patient confidentiality. Besides this, there needs to be transparency and
accountability in model development and deployment. It is important to leverage the models
without algorithmic bias or any sort of inequity.


background image

Another big challenge that remains is that of model interpretability: whereas complex

machine learning models can achieve highly predictive accuracy, the processes through which they
make these decisions may be somewhat incomprehensible. Interpretable models with an ability to
provide transparent explanations of their predictions are what are being developed if trusts are to
be built and clinical adoptions ensured. Furthermore, several limitations indeed need to be
acknowledged in the current study. The sample size could be limited; the generalizability of
findings is restricted because only a certain set of characteristics among the respondents of this
study forms the basis of the observations. The performance of the models may also vary between
different clinical settings and populations.

Future Research Directions

To further enhance the accuracy of predictive algorithms, scholars must consolidate

additional data sources, such as clinical trial data, electronic health records, and real-world
evidence. This incorporation would surely allow researchers to build more robust and informative
models. The integration of real-time patient data into the predictive models facilitates dynamic and
bespoke insights. In summary, continuous monitoring of the clinical status and genomic profile
enables clinicians to adapt treatment plans in real time to optimize outcomes. This can result in
more effective and timely interventions, especially in diseases like cancer that change in a short
period. Moreover, future research should also focus on developing more interpretable and
explainable models through techniques such as feature importance analysis, SHAP values, and
LIME. It is only through the understanding of various factors that go into a model's prediction that
clinicians can confidently trust those recommendations and make informed decisions.

CONCLUSION

This research project aimed to address the challenges of predicting drug sensitivity by

leveraging the GDSC dataset, an extensive resource connecting genomic profiles of cancer cell
lines with their sensitivity to a wide range of anti-cancer drugs. This research's key focus was
identifying robust genomic markers, including any specific mutations, gene expression patterns,
or epigenetic modifications associated with drug sensitivity or resistance. Advanced machine
learning and statistical methods were utilized by the predictive models to analyze complex
relations that may exist between different genomic alterations and their drug sensitivity. The
dataset used for this research project was derived from the Kaggle website. This dataset was
compiled by the research project Genomics of Drug Sensitivity in Cancer collaboration between
the Sanger Institute in the United Kingdom and the Massachusetts General Hospital Cancer Center
in the United States. In their investigation, there was a massive screening of human cancer cell
lines with a wide range of anti-cancer drugs. Data collection was performed by large-scale
screening of diverse anti-cancer drugs against human cancer cell lines of various types. Cell
viability was measured using the Cell-Titer-Glo assay following 72 hours of drug treatment.
Several machine learning models were deployed, namely, Random Forest, Linear Regression, and
XG-Boost, which exhibited specific strengths. Specific performance metrics used included MSE,
RMSE, MAE, and R². As the statistics indicate, among the three models, Random Forest stands
out and performs the best on this dataset across all metrics. A smaller value of MAE, MSE, and


background image

RMSE signifies that it provided the best forecast for the target variable. It also gave the highest R-
squared value. Application of drug sensitivity prediction analysis in cancer can provide an
overview of the mechanisms that underlie both tumor response and resistance by investigating the
model predictions. The proposed predictive models have the potential to make significant impacts
on clinical decision-making in cancer therapy. Predictive models derive informed decisions
regarding a patient's risk of recurrence of disease, their response to certain therapies, and their
prognosis based on complex clinical and genomic data.

References

:

Alipour, S. (2024, August 13).

Genomics of Drug Sensitivity in Cancer (GDSC)

. Kaggle.

https://www.kaggle.com/datasets/samiraalipour/genomics-of-drug-sensitivity-in-cancer-
gdsc

Al Amin, M., Liza, I. A., Hossain, S. F., Hasan, E., Haque, M. M., & Bortty, J. C. (2024).

Predicting and Monitoring Anxiety and Depression: Advanced Machine Learning
Techniques for Mental Health Analysis.

British Journal of Nursing Studies

,

4

(2), 66-75.

Bhowmik, P. K., Miah, M. N. I., Uddin, M. K., Sizan, M. M. H., Pant, L., Islam, M. R., &

Gurung, N. (2024). Advancing Heart Disease Prediction through Machine Learning:
Techniques and Insights for Improved Cardiovascular Health.

British Journal of Nursing

Studies

,

4

(2), 35-50.

Bortty, J. C., Bhowmik, P. K., Reza, S. A., Liza, I. A., Miah, M. N. I., Chowdhury, M. S. R., & Al

uAmin, M. (2024). Optimizing Lung Cancer Risk Prediction with Advanced Machine
Learning Algorithms and Techniques.

Journal of Medical and Health Studies

,

5

(4), 35-48

Chakravarty, D., & Solit, D. B. (2021). Clinical cancer genomic profiling.

Nature Reviews

Genetics

,

22

(8), 483-501.

Chawla, S., Rockstroh, A., Lehman, M., Ratther, E., Jain, A., Anand, A., ... & Sengupta, D.

(2022). Gene expression based inference of cancer drug sensitivity.

Nature

communications

,

13

(1), 5680.

Cobain, E. F., Wu, Y. M., Vats, P., Chugh, R., Worden, F., Smith, D. C., ... & Chinnaiyan, A. M.

(2021). Assessment of clinical benefit of integrative genomic profiling in advanced solid
tumors.

JAMA oncology

,

7

(4), 525-533.

de Jong, J., Cutcutache, I., Page, M., Elmoufti, S., Dilley, C., Fröhlich, H., & Armstrong, M.

(2021). Towards realizing the vision of precision medicine: AI based prediction of
clinical drug response.

Brain

,

144

(6), 1738-1750.

Dutta, S., Sikder, R., Islam, M. R., Al Mukaddim, A., Hider, M. A., & Nasiruddin, M. (2024).

Comparing the Effectiveness of Machine Learning Algorithms in Early Chronic Kidney
Disease Detection.

Journal of Computer Science and Technology Studies

,

6

(4), 77-91.

Hider, M. A., Nasiruddin, M., & Al Mukaddim, A. (2024). Early Disease Detection through

Advanced Machine Learning Techniques: A Comprehensive Analysis and
Implementation in Healthcare Systems.

Revista de Inteligencia Artificial en

Medicina

,

15

(1), 1010-1042.

Hossain, M. S., Rahman, M. K., & Dalim, H. M. (2024). Leveraging AI for Real-Time

Monitoring and Prediction of Environmental Health Hazards: Protecting Public Health in
the USA. Revista de Inteligencia Artificial en Medicina, 15(1), 1117-1145.


background image

Hou, Y. C. C., Yu, H. C., Martin, R., Cirulli, E. T., Schenker-Ahmed, N. M., Hicks, M., ... &

Caskey, C. T. (2020). Precision medicine integrating whole-genome sequencing,
comprehensive metabolomics, and advanced imaging.

Proceedings of the National

Academy of Sciences

,

117

(6), 3053-3062.

Islam, M. Z., Nasiruddin, M., Dutta, S., Sikder, R., Huda, C. B., & Islam, M. R. (2024). A

Comparative Assessment of Machine Learning Algorithms for Detecting and Diagnosing
Breast Cancer.

Journal of Computer Science and Technology Studies

,

6

(2), 121-135.

Lewis, J. E., & Kemp, M. L. (2021). Integration of machine learning and genome-scale

metabolic modeling identifies multi-omics biomarkers for radiation resistance.

Nature

Communications

,

12

(1), 2700.

Li, Y., Umbach, D. M., Krahn, J. M., Shats, I., Li, X., & Li, L. (2021). Predicting tumor response

to drugs based on gene-expression biomarkers of sensitivity learned from cancer cell
lines.

BMC genomics

,

22

, 1-18.

Nasiruddin, M., Dutta, S., Sikder, R., Islam, M. R., Mukaddim, A. A., & Hider, M. A. (2024).

Predicting Heart Failure Survival with Machine Learning: Assessing My Risk.

Journal of

Computer Science and Technology Studies

,

6

(3), 42-55

Prabhod, K. J. (2022). The Role of Machine Learning in Genomic Medicine: Advancements in

Disease Prediction and Treatment.

Journal of Deep Learning in Genomic Data

Analysis

,

2

(1), 1-52.

Pro-AI-Rokibul. (2024).

Machine-Learning-Models-for-Predicting-GDSC-Drug-Response-

Prediction/Model/main.ipynb at main · proAIrokibul/Machine-Learning-Models-for-
Predicting-GDSC-Drug-Response-Prediction

. GitHub.

https://github.com/proAIrokibul/Machine-Learning-Models-for-Predicting-GDSC-Drug-
Response-Prediction/blob/main/Model/main.ipynb

Quazi, S. (2022). Artificial intelligence and machine learning in precision and genomic

medicine.

Medical Oncology

,

39

(8), 120.

Rahman, A., Karmakar, M., & Debnath, P. (2023). Predictive Analytics for Healthcare:

Improving Patient Outcomes in the US through Machine Learning.

Revista de

Inteligencia Artificial en Medicina

,

14

(1), 595-624

Thirunavukarasu, R., Gnanasambandan, R., Gopikrishnan, M., & Palanisamy, V. (2022).

Towards computational solutions for precision medicine based big data healthcare system
using deep learning models: A review.

Computers in Biology and Medicine

,

149

, 106020.

Библиографические ссылки

Alipour, S. (2024, August 13). Genomics of Drug Sensitivity in Cancer (GDSC). Kaggle.

https://www.kaggle.com/datasets/samiraalipour/genomics-of-drug-sensitivity-in-cancer-gdsc

Al Amin, M„ Liza, I. A., Hossain, S. E, Hasan, E., Haque, M. M„ & Bortty, J. C. (2024). Predicting and Monitoring Anxiety and Depression: Advanced Machine Learning Techniques for Mental Health Analysis. British Journal of Nursing Studies, 4(2), 66-75.

Bhowmik, P. K., Miah, M. N. I., Uddin, M. K.., Sizan, M. M. H., Pant, L., Islam, M. R., & Gurung, N. (2024). Advancing Heart Disease Prediction through Machine Learning: Techniques and Insights for Improved Cardiovascular Health. British Journal of Nursing Studies, 4(2), 35-50.

Bortty, J. C., Bhowmik, P. K., Reza, S. A., Liza, I. A., Miah, M. N. I., Chowdhury, M. S. R., & Al uAmin, M. (2024). Optimizing Lung Cancer Risk Prediction with Advanced Machine Learning Algorithms and Techniques. Journal of Medical and Health Studies, 5(4), 35-48

Chakravarty, D., & Solit, D. B. (2021). Clinical cancer genomic profiling. Nature Reviews Genetics, 22(8), 483-501.

Chawla, S., Rockstroh, A., Lehman, M., Ratther, E., Jain, A., Anand, A.,... & Sengupta, D. (2022). Gene expression based inference of cancer drug sensitivity. Nature communications, 13(), 5680.

Cobain, E. E, Wu, Y. M., Vats, R, Chugh, R„ Worden, E, Smith, D. C.,... & Chinnaiyan, A. M. (2021). Assessment of clinical benefit of integrative genomic profiling in advanced solid tumors. JAMA oncology, 7(4), 525-533.

de Jong, J., Cutcutache, I., Page, M., Elmoufti, S„ Dilley, C„ Frohlich, H„ & Armstrong, M. (2021). Towards realizing the vision of precision medicine: Al based prediction of clinical drug response. Brain, 144(6), 1738-1750.

Dutta, S„ Sikder, R., Islam, M. R., Al Mukaddim, A., Hider, M. A., & Nasiruddin, M. (2024). Comparing the Effectiveness of Machine Learning Algorithms in Early Chronic Kidney Disease Detection. Journal of Computer Science and Technology Studies, 6(4), 77-91.

Hider, M. A., Nasiruddin, M„ & Al Mukaddim, A. (2024). Early Disease Detection through Advanced Machine Learning Techniques: A Comprehensive Analysis and Implementation in Healthcare Systems. Revista de Inteligencia Artificial en Medicina, /5(1), 1010-1042.

Hossain, M. S., Rahman, M. K., & Dalim, H. M. (2024). Leveraging Al for Real-Time Monitoring and Predietion of Environmental Health Hazards: Protecting Public Health in the USA. Revista de Inteligencia Artificial cn Medicina, 15(1), 1117-1145.

Hou, Y. С. C., Yu, H. C„ Martin, R., Cirulli, E. T„ Schenker-Ahmed, N. M„ Hicks, M.,... & Caskey, С. T. (2020). Precision medicine integrating whole-genome sequencing, comprehensive metabolomics, and advanced imaging. Proceedings of the National Academy of Sciences, 117(6), 3053-3062.

Islam, M. Z., Nasiruddin, M„ Dutta, S., Sikder, R., Huda, С. B., & Islam, M. R. (2024). A Comparative Assessment of Machine Learning Algorithms for Detecting and Diagnosing Breast Cancer. Journal of Computer Science and Technology Studies, 6(2), 121-135.

Lewis, J. E., & Kemp, M. L. (2021). Integration of machine learning and genome-scale metabolic modeling identifies multi-omics biomarkers for radiation resistance. Nature Communications, /2(1), 2700.

Li, Y„ Umbach, D. M., Krahn, J. M., Shats, L, Li, X., & Li, L. (2021). Predicting tumor response to drugs based on gene-expression biomarkers of sensitivity learned from cancer cell lines. BMC genomics, 22, 1-18.

Nasiruddin, M„ Dutta, S., Sikder, R., Islam, M. R., Mukaddim, A. A., & Hider, M. A. (2024).

Predicting Heart Failure Survival with Machine Learning: Assessing My Risk. Journal of Computer Science and Technology Studies, 6(3), 42-55

Prabhod, K. J. (2022). The Role of Machine Learning in Genomic Medicine: Advancements in Disease Prediction and Treatment. Journal of Deep Learning in Genomic Data Analysis, 2(1), 1-52.

Pro-AI-Rokibul. (2024). Machine-Learning-Models-for-Predicting-GDSC-Drug-Response-Prediction/Model/main. ipynb at main • proAIrokibul/Machine-Learning-Models-for-Predicting-GDSC-Drug-Response-Prediction. GitHub.

https://github.com/proAIrokibul/Machine-Learning-Models-for-Predicting-GDSC-Drug-Response-Prediction/blob/main/Model/main. ipynb

Quazi, S. (2022). Artificial intelligence and machine learning in precision and genomic medicine. Medical Oncology, 39(8), 120.

Rahman, A., Karmakar, M., & Debnath, P. (2023). Predictive Analytics for Healthcare: Improving Patient Outcomes in the US through Machine Learning. Revista de Inteligencia Artificial en Medicina, 14(), 595-624

Thirunavukarasu, R„ Gnanasambandan, R., Gopikrishnan, M„ & Palanisamy, V. (2022).

Towards computational solutions for precision medicine based big data healthcare system using deep learning models: A review. Computers in Biology and Medicine, 149, 106020.