Quantum Gradient Optimized Drug Repurposing Prototype for
Omics Data
Don Roosan
1
, Saif Nirzhor
2
, Rubayat Khan
3
and Fahmida Hai
4
1
School of Engineering and Computational Sciences, Merrimack College, North Andover, U.S.A.
2
University of Texas Southwestern Medical Center, Dallas, U.S.A.
3
University of Nebraska Medical Center, Omaha, U.S.A.
4
Tekurai Inc., San Antonio, U.S.A.
Keywords: Quantum Computing, Drug Repurposing, Massive Genomics Data, Large Language Models, Quantum
Gradient.
Abstract: This paper presents a novel quantum-enhanced prototype for drug repurposing and addresses the challenge of
managing massive genomics data in precision medicine. Leveraging cutting-edge quantum server
architectures, we integrated quantum-inspired feature extraction with large language model (LLM)–-based
analytics and unified high-dimensional omics datasets and textual corpora for faster and more accurate
therapeutic insights. Applying Synthetic Minority Over-sampling Technique (SMOTE) to balance
underrepresented cancer subtypes and multi-omics sources such as TCGA and LINCS, the pipeline generated
refined embeddings through quantum principal component analysis (QPCA). These embeddings drove an
LLM trained on biomedical texts and clinical notes, generating drug recommendations with improved
predicted efficacy and safety profiles. Combining quantum computing with LLM outperformed classical
PCA-based approaches in accuracy, F1 score, and area under the ROC curve. Our prototype highlights the
potential of harnessing quantum computing and next-generation servers for scalable, explainable, and timely
drug repurposing in modern healthcare.
1 INTRODUCTION
Drug repositioning, also known as drug repurposing,
has emerged as a critical strategy for accelerating
therapeutic innovation within the modern healthcare
environment. The conventional drug discovery
process, typically spanning more than a decade,
demands an immense financial outlay (Corsello et al.,
2017; Park, 2019). Moreover, even after preliminary
regulatory approvals, many candidate drugs fail in
subsequent clinical trial phases, resulting in
significant resource loss. Repurposing approved
drugs or late-stage clinical candidates offers a much
more efficient path. These compounds already come
with known safety profiles and pharmacokinetic
characteristics, allowing researchers to skip multiple
preclinical steps, thereby saving both time and
money. This efficiency is especially attractive when
confronting urgent or emerging health crises—such
as newly identified viral pathogens, widespread
cancers, or rare diseases with limited treatment
options—where speed can be a decisive factor. One
of the chief reasons drug repurposing has garnered
attention is the ability to sidestep the most daunting
and time-intensive stages of drug development.
Under standard protocols, discovering and validating
a novel compound involves extensive preclinical
testing to gauge toxicity, clinical complexity dosing
parameters, and efficacy in animal models before it
can even proceed to trials in humans (Islam et al.,
2014; Islam, Mayer, et al., 2016; Islam, Weir, et al.,
2016a, 2016b). These steps alone can consume years
and require substantial financial support. By turning
to substances already verified as safe for human use,
scientists can concentrate more on efficacy for a
novel indication, substantially reducing the overall
timeline. This approach also heightens the likelihood
of success in advanced clinical trials, as the critical
factor of human safety has been substantially
addressed.
Speed is paramount in public health crises,
making repurposing strategies particularly relevant.
When rapid drug deployment is critical, such as
during a pandemic, compounds that are already
Roosan, D., Nirzhor, S., Khan, R., Hai and F.
Quantum Gradient Optimized Drug Repurposing Prototype for Omics Data.
DOI: 10.5220/0013524900003967
In Proceedings of the 14th Inter national Conference on Data Science, Technology and Applications (DATA 2025), pages 465-472
ISBN: 978-989-758-758-0; ISSN: 2184-285X
Copyright © 2025 by Paper published under CC license (CC BY-NC-ND 4.0)
465
approved—or very close to approval—can be
mobilized faster. If a known medication reveals the
potential to inhibit an emergent virus or alleviate
severe symptoms, it stands a significantly higher
chance of quickly reaching clinical use(Graves et al.,
2018; Islam et al., 2015). Beyond pandemic
scenarios, this model can extend to diseases where no
existing interventions are available, including
neglected tropical diseases or rare genetic disorders,
illustrating how the timeliness afforded by drug
repositioning can address unmet medical
needs(Tukur et al., 2023).
Along with saving time and resources, drug
repurposing is conceptually powerful. A drug
designed for one purpose may act on multiple
biological pathways, broadening its therapeutic
impact. Advances in fields such as genomics and
proteomics have deepened our understanding of how
diseases can share overlapping molecular
mechanisms, supporting a rationale for exploring the
off-label application of known drugs. As knowledge
about intricate disease networks continues to grow,
the argument for systematically examining
alternative uses of existing drugs becomes even more
convincing (Roosan et al., 2019). This approach
effectively acts as a shortcut, delivering novel
treatments to patients more rapidly than de novo drug
discovery efforts typically allow.
Drug repurposing, using medications for new
indications, has a long history, exemplified by
Aspirin (initially for pain, later for heart conditions)
and thalidomide (sedative, later for leprosy and
cancer). Modern methods leverage data-driven
approaches, including bioinformatics and machine
learning, to analyze molecular and clinical datasets
for new drug applications (Roosan et al., 2021;
Roosan, Wu, Tran, et al., 2022; Deng et al., 2022).
However, complex diseases like cancer and diabetes,
involving intricate gene-protein-environment
interactions, and polypharmacology (drugs affecting
multiple mechanisms) make repurposing data-
intensive. Big data from genomics, proteomics, and
electronic medical records offers opportunities but
poses integration challenges due to diverse data types
and high dimensionality (Li et al., 2021; Roosan,
Hwang, et al., 2020; Sammani et al., 2019).
Traditional computational tools struggle with the
"curse of dimensionality," necessitating advanced
analytical methods for biomedical data (Cao et al.,
2011; Roosan et al., 2017). Quantum computing
promises to revolutionize drug repurposing by using
qubits’ superposition and entanglement to efficiently
analyze large datasets (D. Roosan et al., 2024; Doga
& et al., 2024; J. Yang et al., 2024). Though limited
by qubit count and error rates, quantum-inspired
algorithms like quantum kernel methods and
principal component analysis uncover hidden
patterns in biomedical data (Jeyaraman et al., n.d.;
Sung et al., 2018). Applications include precise
molecular modeling for protein-ligand docking and
combinatorial optimization for drug-disease pairings
(D. Roosan et al., 2024; Pandey et al., 2024). LLMs
excel at processing unstructured text and multimodal
data, revealing connections between diseases,
biomarkers, and drugs (Wu et al., 2012; R. Yang et
al., 2023). Challenges include misinformation, biases,
and transparency issues. Quantum-enhanced feature
extraction paired with LLMs could streamline drug
repurposing by tackling high-dimensional data and
interpreting findings (Islam et al., 2014; Itri & Patel,
2018). Disease complexity and big data demand
advanced computational strategies beyond heuristics,
leveraging AI-driven tools for healthcare insights.
Blockchain technology enhances secure,
transparent healthcare data sharing for drug
repurposing by storing immutable records of patient
consents and data access (Dhillon et al., 2017;
Roosan, Wu, Tatla, et al., 2022). In pandemics, AI-
driven analytics, augmented by quantum-inspired
methods, expedite identifying existing drugs for new
pathogens (Challen et al., 2019; Roosan, Chok, et al.,
2020; Roosan et al., 2023). A quantum-enhanced,
large language model (LLM)-based system could
analyze large datasets—from genomic profiles to
clinical observations—using quantum-inspired
feature extraction and LLM interpretation to suggest
repurposed drug candidates with rationales. The
primary goal is to develop and validate a prototype of
this system, focusing on patient-specific data analysis
and interpretable recommendations using high-
dimensional genomic and clinical data.
2 METHOD
2.1 Dataset Preparation
This study utilized a multi-faceted approach to data
collection and integration, aiming to create a robust
foundation for a quantum-enhanced, large language
model (LLM)-driven drug repurposing system. The
dataset encompassed clinical records, synthetic
patient cohorts, and three major omics repositories,
all meticulously harmonized to ensure consistency in
format, terminology, and quality.
DATA 2025 - 14th International Conference on Data Science, Technology and Applications
466
2.1.1 Clinical Data
The MIMIC-III database, a comprehensive collection
of de-identified critical care patient data, served as the
primary source of clinical information (D. Clifford et
al., 2009). The database includes a wealth of data
points, such as patient demographics, vital signs, lab
results, medication records, and outcomes. For this
study, the data extraction focused on oncology-
related cases. The selection criteria prioritized
patients with documented cancer diagnoses, cancer-
related medication records, and sufficient laboratory
data to enable in-depth exploration of their disease
status. Standard protocols for de-identification and
privacy compliance were strictly adhered to
throughout the data handling process. The MIMIC-III
database includes information from over 40,000
patients admitted to critical care units at the Beth
Israel Deaconess Medical Center between 2001 and
2012.
2.1.2 Synthetic Cohort Generation
While MIMIC-III offers a broad range of patient
records, certain cancer subtypes and demographic
groups were underrepresented. To mitigate potential
biases arising from class imbalance, the Synthetic
Minority Over-sampling Technique (SMOTE) was
employed. SMOTE generated 60 synthetic patient
records, effectively balancing the representation of
prevalent or majority classes with those representing
rare or less frequently documented conditions. Each
synthetic patient entry included key features such as
demographic data (e.g., age, sex), lab results (e.g.,
complete blood counts, serum chemistry panels), vital
signs (e.g., systolic and diastolic blood pressure), and
clinical outcome indicators (e.g., survival or
readmission rates). This process ensured a more
uniform representation of diseases and disease stages,
resulting in a more robust training set for machine
learning algorithms. For instance, in the original
MIMIC-III dataset, African American patients
comprised approximately 9% of the total, while
Hispanic patients made up around 3%. After applying
SMOTE, the representation of these groups in the
synthetic cohort was increased to approximately 15%
each, providing a more balanced dataset for training
the model (Chawla et al., 2002).
2.2 Omics Data
In addition to clinical features, the system
incorporated molecular-level information to identify
biological patterns and potential drug repurposing
opportunities.
2.2.1 The Cancer Genome Atlas (TCGA)
RNA-sequencing data for breast cancer (BRCA)
samples were obtained from the TCGA portal. Strict
inclusion criteria ensured that only high-quality gene
expression profiles with robust clinical annotations,
such as tumor stage, lymph node involvement, and
other pathological features, were included. Each
sample's raw data was normalized using established
protocols, and the resulting gene expression matrices
were used for downstream analyses. The TCGA
database contains genomic data from over 11,000
patients across 33 different cancer types. For this
study, the breast cancer (BRCA) subset, which
includes data from approximately 1,100 patients, was
utilized (Tomczak et al., 2015).
2.2.2 Gene Expression Omnibus (GEO)
To further refine and validate cancer-specific gene
expression trends, the GSE2034 dataset, a well-
curated dataset relevant to breast cancer prognosis,
was integrated. This dataset contained microarray-
based expression values, which were meticulously
normalized and mapped to the same gene symbols
used in the TCGA-BRCA subset. The integration of
microarray data from GEO helped address potential
biases arising from reliance on a single technology or
population. The GSE2034 dataset includes gene
expression data from 286 breast cancer patients,
providing a valuable resource for validating findings
from the TCGA data (Barrett et al., 2013).
2.2.3 Library of Integrated Network-Based
Cellular Signatures (LINCS)
The LINCS L1000 dataset provided crucial
information on how various small-molecule
compounds affect gene expression across diverse cell
lines. The focus was on expression signatures that
captured drug-induced upregulation or
downregulation of genes relevant to cancer pathways.
This resource was essential for training the model to
link unique patient gene expression patterns to
possible therapeutic compounds, highlighting
existing drugs that could be repurposed based on their
cellular signatures. The LINCS L1000 dataset
contains gene expression profiles from over 1 million
experiments, measuring the effects of approximately
20,000 small-molecule compounds on various cell
lines (Duan et al., 2016).
Quantum Gradient Optimized Drug Repurposing Prototype for Omics Data
467
2.3 Data Pre-Processing
Data from these multiple sources underwent extensive
processing to ensure standardization and
comparability across clinical and molecular domains.
All patient records, both downloaded and
synthetically generated, underwent thorough quality
checks. Missing values in the clinical data were
imputed using mean or median values, depending on
the distribution of each feature. Outlier detection was
performed by setting interquartile range (IQR)
thresholds, removing samples with extreme values
that could skew the training process. Gene expression
matrices, from both RNA-seq and microarray sources,
were subjected to low-expression filtering,
eliminating genes that lacked sufficient read counts in
most samples.
Feature engineering began with the harmonization
of gene symbols across TCGA-BRCA, GSE2034, and
L1000 data. A targeted approach to feature selection
identified genes with the greatest variance across
disease subtypes, known cancer driver genes, and
genes encoding enzymes relevant to drug metabolism
(e.g., certain cytochrome P450 isoforms).
Dimensionality reduction was initiated through
classical Principal Component Analysis (PCA).
However, the core innovation involved feeding these
partially reduced features into a quantum-inspired
algorithm for further compression, preserving non-
linear relationships that PCA might overlook. In a
manner akin to the data-integration strategies
employed in nutrigenomics—where RNA and DNA
testing illuminate gene-environment interactions we
applied quantum feature mapping within our GNN
framework to enrich molecular data representations.
Recent advances in healthcare informatics
demonstrate how data visualization techniques, such
as heatmaps, can streamline the processing of
complex, unstructured data from electronic health
records (EHRs). Standardizing data is essential for
improving health information exchange and
interoperability, although it is often overlooked in
system-level implementations.
2.4 Quantum Enhanced LLM-Based
Drug Repurposing Model
After preparing cleaned datasets and curated features,
a framework combined quantum-inspired feature
extraction—using simulated Quantum Principal
Component Analysis (QPCA) and quantum kernel
methods due to hardware limitations—with a
transformer-based language model (LLM) for drug
prediction. Gradient-based optimization refined
embeddings. An SVM with a linear kernel classified
drug matches using MIMIC-III and synthetic
SMOTE-generated data, with specified training,
testing, and validation splits. The core system
integrated quantum-enriched embeddings with an
LLM fine-tuned on drug-disease relationships,
biomedical literature, and patient metadata. The
pipeline normalized raw data, reduced dimensions via
classical PCA and quantum transformations, used the
LLM to predict drug efficacy, and ranked candidates
by efficacy and safety. Hyperparameters, code, and
synthetic data are publicly available.To support
reproducibility, we included hyperparameter settings
and made the code and synthetic data publicly
available, addressing the experimentation discussion’s
prior lack of detail.
3 RESULTS
3.1 Model Performance
The quantum-enhanced feature extraction
demonstrated robust gains relative to purely classical
approaches. To quantitatively evaluate these
improvements, we computed standard classification
metrics—accuracy, precision, recall, F1-score, and the
area under the ROC curve (AUC)—for predicting a
successful drug match. A total of 1,200 labeled
instances (synthetic patients with known best treatment
outcomes or prospective matches) were used for
validation. The quantum-inspired transformations
consistently outperformed classical PCA, yielding a
higher F1-score by an average of 8% across multiple
runs. Notably, some samples with subtle gene
expression shifts only achieved clinically meaningful
matches when the quantum kernel transformations
Figure 1: Comparative performance of classical PCA-based
dimensionality reduction versus quantum-enhanced feature
engineering.
DATA 2025 - 14th International Conference on Data Science, Technology and Applications
468
were included, underscoring the sensitivity of this
approach to nuanced genetic variation. The quantum-
based plot reveals tighter clustering among patients
with similar clinical and and molecular profiles,
suggesting an improved capacity for separating
responders from non-responders.
To supplement these visual indicators, the
summary of performance metrics is shown in Table
1, comparing the average performance standard
deviation) across five cross-validation folds.
Table 1: Cross-Validation Performance Comparison of
Classical vs. Quantum Approaches.
Method
Accuracy
(%)
Precision
(%)
Recall
(%)
F1-
Score
(%)
AUC
Classica
l PCA
82.3 ±
2.1
80.7 ± 1.9
78.9 ±
2.6
79.8
± 2.4
0.84
±
0.03
Quantu
m-
Enhance
d
88.5 ±
1.8
86.2 ± 2.0
84.7 ±
2.1
85.4
± 1.9
0.90
±
0.02
The table includes accuracy, precision, recall, F1-
score, and AUC, demonstrating consistent gains in all
metrics under the quantum-enhanced setting.To
further highlight the dimensionality reduction aspect,
Figure 2 presents a two-dimensional t-SNE
projection of patient embeddings. The upper panel
(Figure 2A) shows the clustering using only classical
PCA, whereas the lower panel (Figure 2B) overlays
the quantum-transformed embeddings on the same
manifold. We enhanced clustering evaluation by
Figure 2. A two-dimensional t-SNE projection of
patient embeddings, comparing classical PCA-based
clustering (2A) with quantum-transformed
embeddings (2B) on the same manifold.
Incorporating robust metrics such as a silhouette
index of 0.65 for quantum-enhanced embeddings (vs.
0.52 for classical PCA), acknowledging synthetic
data limitations and the need for real clinical
validation. To substantiate efficacy, we compared our
quantum-enhanced model to classical PCA-based
methods on the TCGA-BRCA dataset, achieving a
10% F1-score improvement, with plans to benchmark
against additional state-of-the-art solutions in future
work.
A final summary of representative drug
recommendations is provided in Table 2,
documenting sample outputs for three synthetic
patients with varying clinical statuses. Each row
indicates top drugs, predicted efficacy scores, and
relevant gene targets implicated in the drug match.
Table 2: Excerpt of Drug Recommendations for Three
Synthetic Patients.
Patient
ID
Stage
Top
Recommended
Drug
Predicted
Efficacy
(%)
Key Gene
Targets
SYN-
01
II Palbociclib 78
CDK4,
CDK6
SYN-
24
III Tamoxifen 82
ESR1,
ESR2
SYN-
47
IIIB Sorafenib 74
RAF,
VEGFR,
PDGFR
This table highlights a range of therapy classes—
hormone modulators, kinase inhibitors, and multi-
target agents—all emerging as candidates from the
pipeline’s predictions based on individual patient
molecular and clinical characteristics. Integrating the
quantum-inspired transformations appears to sharpen
distinctions between viable and less-appropriate
options, potentially accelerating the pace of drug
discovery and repurposing for oncology care. These
results are consistent with earlier research
demonstrating that integrating AI techniques can
significantly improve data analysis and decision-
making
3.2 Synthetic Cohort
For the 60-patient synthetic cohort generated via
SMOTE, each individual’s record was processed
through the pipeline to derive recommended
treatments. These synthetic cases were diverse in
terms of disease stages, comorbidity indices, and
molecular profiles. The quantum-enhanced approach
proved particularly valuable in identifying relevant
kinase inhibitors and hormone modulators for
patients mimicking more advanced stages of breast
cancer. Many of these suggestions aligned with
known FDA-approved drugs in related contexts,
thereby highlighting the potential for repurposing in
real-world scenarios.
Quantum Gradient Optimized Drug Repurposing Prototype for Omics Data
469
4 DISCUSSIONS
Our prototype revolutionizes clinical decision-
making by integrating high-dimensional omics data
with clinical records. Unlike traditional drug
repurposing, which relies on slow literature reviews
and outdated machine learning, our quantum-inspired
approach excels at uncovering gene-protein-disease
relationships. Processed by a fine-tuned LLM, it
delivers clear, interpretable drug recommendations,
aiding clinicians in complex cases where standard
treatments fail (Roosan, 2023; Roosan et al., 2023;
Roosan, Roosan, Kim, et al., 2022). The LLM
enhances transparency with explanations linking
recommendations to gene-drug interactions,
validated by synthetic data, though real-world trials
are needed. Its user-friendly interface lets clinicians
explore reasoning from literature, omics, and patient
data, reducing "black-box" skepticism and boosting
trust. Public awareness of off-label treatments,
supported by LLM-generated summaries, can drive
advocacy and trial enrollment, fostering trust in data
sharing. In policy, our quantum-enhanced LLM aids
lawmakers by assessing drug viability, balancing
innovation, safety, and costs. Explainable AI
translates molecular insights into policy-friendly
narratives, speeding up therapy adoption. The
prototype supports future multimodal LLMs,
integrating voice, images, and video for a holistic
patient view, improving equity and precision.
Challenges include quantum hardware costs, LLM
training demands, and integration complexities. Data
biases and latency need addressing, but investment in
quantum research and LLM efficiency can overcome
these. Limitations include hardware constraints, data
requirements, and the need for diverse clinical
validation. Future work will refine the system.
5 CONCLUSIONS
The prototype presented in this study offers a tangible
advancement in drug repurposing by combining
quantum-inspired feature extraction with LLM–based
analytics. By orchestrating high-dimensional omics
datasets—such as RNA-seq and microarray gene
expression profiles—with detailed clinical
information, the system demonstrates a clear
capability to prioritize potential therapies for diverse
patient populations. Unlike conventional machine
learning methods that struggle to handle complex and
expansive data, the quantum-enhanced approach
excels at discerning subtle patterns in gene
expression, ultimately improving classification
metrics such as accuracy, F1-score, and area under
the ROC curve. The pipeline’s ability to integrate
QPCA with LLM-driven interpretation highlights the
potential for scalable, explainable, and timely
solutions in modern healthcare. Though current
hardware limitations and computational demands
pose practical challenges, ongoing innovations in
quantum simulators and AI architectures will likely
reduce operating costs and further streamline this
approach.
ACKNOWLEDGEMENTS
We are grateful to Merrimack College for support.
REFERENCES
Barrett, T., Wilhite, S. E., Ledoux, P., Evangelista, C., Kim,
I. F., Tomashevsky, M., Marshall, K. A., Phillippy, K.
H., Sherman, P. M., Holko, M., Yefanov, A., Lee, H.,
Zhang, N., Robertson, C. L., Serova, N., Davis, S., &
Soboleva, A. (2013). NCBI GEO: Archive for
functional genomics data sets—update. Nucleic Acids
Research, 41(D1), D991–D995. https://doi.org/10.10
93/nar/gks1193
Cao, N., Gotz, D., Sun, J., & Qu, H. (2011). Dicon:
Interactive visual analysis of multidimensional clusters.
Visualization and Computer Graphics, IEEE
Transactions On, 17(12), Article 12.
Challen, R., Denny, J., Pitt, M., Gompels, L., Edwards, T.,
& Tsaneva-Atanasova, K. (2019). Artificial
intelligence, bias and clinical safety. BMJ Quality &
Safety, 28(3), 231–237.
Chawla, N. V., Bowyer, K. W., Hall, L. O., & Kegelmeyer,
W. P. (2002). SMOTE: Synthetic Minority Over-
sampling Technique. Journal of Artificial Intelligence
Research, 16, 321–357. https://doi.org/10.1613/jair.953
Corsello, S. M., Bittker, J. A., Liu, Z., Gould, J., McCarren,
P., Hirschman, J. E., Johnston, S. E., Vrcic, A., Wong,
B., & Khan, M. (2017). The Drug Repurposing Hub: A
next-generation drug library and information resource.
Nature Medicine, 23(4), 405–408. https://www.nature.
com/articles/nm.4306
Clifford, G. D., Scott, D., & Villarroel, M. (2009). User
guide and documentation for the MIMIC II database.
Roosan, D., Chok, J., Li, Y., & Khou T. (2024). Utilizing
Quantum Computing-based Large Language
Transformer Models to Identify Social Determinants of
Health from Electronic Health Records. 2024
International Conference on Electrical, Computer and
Energy Technologies (ICECET, 1–6. https://doi.org/
10.1109/ICECET61485.2024.10698600
Deng, J., Yang, Z., Ojima, I., Samaras, D., & Wang, F.
(2022). Artificial intelligence in drug discovery:
DATA 2025 - 14th International Conference on Data Science, Technology and Applications
470
Applications and techniques. Briefings in
Bioinformatics, 23(1), bbab430. https://doi.org/10.10
93/bib/bbab430
Dhillon, V., Metcalf, D., & Hooper, M. (2017). Blockchain
in Health Care. In Blockchain Enabled Applications
(pp. 125–138). Apress, Berkeley, CA. https://link.
springer.com/chapter/10.1007/978-1-4842-3081-7_9
Doga, H. & et al. (2024). Towards quantum computing for
clinical trial design and optimization: A perspective on
new opportunities and challenges. https://doi.org/
10.48550/arXiv.2404.13113
Duan, Q., Reid, S. P., Clark, N. R., Wang, Z., Fernandez,
N. F., Rouillard, A. D., Readhead, B., Tritsch, S. R.,
Hodos, R., Hafner, M., Niepel, M., Sorger, P. K.,
Dudley, J. T., Bavari, S., Panchal, R. G., & Ma’ayan,
A. (2016). L1000CDS2: LINCS L1000 characteristic
direction signatures search engine. Npj Systems Biology
and Applications, 2(1), 16015. https://doi.org/1
0.1038/npjsba.2016.15
Graves, K. D., Hall, M. J., & Tercyak, K. P. (2018).
Introduction to the Special Issue on Clinical and Public
Health Genomics: Opportunities for translational
behavioral medicine research, practice, and policy.
Translational Behavioral Medicine, 8(1), Article 1.
https://doi.org/10.1093/tbm/ibx068
Islam, R., Mayer, J., & Clutter, J. (2016). Supporting novice
clinicians cognitive strategies: System design
perspective. IEEE-EMBS International Conference on
Biomedical and Health Informatics. IEEE-EMBS
International Conference on Biomedical and Health
Informatics, 2016, 509. https://doi.org/10.1109/BHI.
2016.7455946
Islam, R., Weir, C., & Del Fiol, G. (2016a). Clinical
Complexity in Medicine: A Measurement Model of
Task and Patient Complexity. Methods of Information
in Medicine, 55(01), 14–22. https://doi.org/
10.3414/ME15-01-0031
Islam, R., Weir, C., & Del Fiol, G. (2016b). Clinical
Complexity in Medicine: A Measurement Model of
Task and Patient Complexity. Methods of Information
in Medicine, 55(1), Article 1. https://doi.org/
10.3414/ME15-01-0031
Islam, R., Weir, C., & Fiol, G. D. (2014). Heuristics in
Managing Complex Clinical Decision Tasks in Experts’
Decision Making. 186–193. https://doi.org/10.1109/
ICHI.2014.32
Islam, R., Weir, C. R., Jones, M., Del Fiol, G., & Samore,
M. H. (2015). Understanding complex clinical
reasoning in infectious diseases for improving clinical
decision support design. BMC Medical Informatics and
Decision Making, 15(1), 101. https://doi.org/10.1186/
s12911-015-0221-z
Itri, J. N., & Patel, S. H. (2018). Heuristics and Cognitive
Error in Medical Imaging. American Journal of
Roentgenology, 210(5), Article 5. https://doi.org/
10.2214/AJR.17.18907
Jeyaraman, N., Jeyaraman, M., Yadav, S.,
Ramasubramanian, S., & Balaji, S. (n.d.).
Revolutionizing Healthcare: The Emerging Role of
Quantum Computing in Enhancing Medical
Technology and Treatment. Cureus, 16(8), e67486.
https://doi.org/10.7759/cureus.67486
Li, Y., Duche, A., Sayer, M. R., Roosan, D., Khalafalla, F.
G., Ostrom, R. S., Totonchy, J., & Roosan, M. R.
(2021). SARS-CoV-2 early infection signature
identified potential key infection mechanisms and drug
targets. BMC Genomics, 22(1), Article 1.
https://doi.org/10.1186/s12864-021-07433-4
Pandey, S., Bhushan, B., & Hameed, A. A. (2024).
Securing Healthcare 5.0: Zero-Knowledge Proof (ZKP)
and Post Quantum Cryptography (PQC) Solutions for
Medical Data Security. In C. K. K. Reddy, T. Sithole,
M. Ouaissa, Ö. ÖZER, & M. M. Hanafiah (Eds.), Soft
Computing in Industry 5.0 for Sustainability (pp. 339–
355). Springer Nature Switzerland. https://doi.org/
10.1007/978-3-031-69336-6_15
Park, K. (2019). A review of computational drug
repurposing. Translational and Clinical
Pharmacology, 27(2), 59–63. https://synapse.korea
med.org/articles/1130297
Roosan, D. (2023). Augmented Reality and Artificial
Intelligence: Applications in Pharmacy. In V.
Geroimenko (Ed.), Augmented Reality and Artificial
Intelligence: The Fusion of Advanced Technologies
(pp. 227–243). Springer Nature Switzerland.
https://doi.org/10.1007/978-3-031-27166-3_13
Roosan, D., Chok, J., Karim, M., Law, A. V., Baskys, A.,
Hwang, A., & Roosan, M. R. (2020). Artificial
Intelligence–Powered Smartphone App to Facilitate
Medication Adherence: Protocol for a Human Factors
Design Study. JMIR Res Protoc, 9(11), Article 11.
https://doi.org/10.2196/21659
Roosan, D., Hwang, A., Law, A. V., Chok, J., & Roosan,
M. R. (2020). The inclusion of health data standards in
the implementation of pharmacogenomics systems: A
scoping review. Pharmacogenomics, 21(16), 1191–
1202. https://doi.org/10.2217/pgs-2020-0066
Roosan, D., Hwang, A., & Roosan, M. R. (2021).
Pharmacogenomics cascade testing (PhaCT): A novel
approach for preemptive pharmacogenomics testing to
optimize medication therapy. The Pharmacogenomics
Journal, 21(1), 1–7. https://doi.org/10.1038/s41397-
020-00182-9
Roosan, D., Law, A. V., Karim, M., & Roosan, M. (2019).
Improving Team-Based Decision Making Using Data
Analytics and Informatics: Protocol for a Collaborative
Decision Support Design. JMIR Res Protoc, 8(11),
Article 11. https://doi.org/10.2196/16047
Roosan, D., Padua, P., Khan, R., Khan, H., Verzosa, C., &
Wu, Y. (2023). Effectiveness of ChatGPT in clinical
pharmacy and the role of artificial intelligence in
medication therapy management. Journal of the
American Pharmacists Association. https://doi.org/
10.1016/j.japh.2023.11.023
Roosan, D., Roosan, M. R., Kim, S., Law, A. V., & Sanine,
C. (2022). Applying Artificial Intelligence to create risk
stratification visualization for underserved patients to
improve population health in a community health
setting. https://doi.org/10.21203/rs.3.rs-1650806/v1
Quantum Gradient Optimized Drug Repurposing Prototype for Omics Data
471
Roosan, D., Weir, C., Samore, M., Jones, M., Rahman, M.,
Stoddard, G. J., & Del Fiol, G. (2017). Identifying
Complexity in Infectious Diseases Inpatient Settings:
An Observation Study. Journal of Biomedical
Informatics, 71 Suppl, S13–S21. https://doi.org/
10.1016/j.jbi.2016.10.018
Roosan, D., Wu, Y., Tatla, V., Li, Y., Kugler, A., Chok, J.,
& Roosan, M. R. (2022). Framework to enable
pharmacist access to health care data using Blockchain
technology and artificial intelligence. Journal of the
American Pharmacists Association, 62(4), 1124–1132.
https://doi.org/10.1016/j.japh.2022.02.018
Roosan, D., Wu, Y., Tran, M., Huang, Y., Baskys, A., &
Roosan, M. R. (2022). Opportunities to integrate
nutrigenomics into clinical practice and patient
counseling. European Journal of Clinical Nutrition.
https://doi.org/10.1038/s41430-022-01146-x
Sammani, A., Jansen, M., Linschoten, M., Bagheri, A., De
Jonge, N., Kirkels, H., Van Laake, L. W., Vink, A., Van
Tintelen, J. P., Dooijes, D., Te Riele, A. S. J. M.,
Harakalova, M., Baas, A. F., & Asselbergs, F. W.
(2019). UNRAVEL: Big data analytics research data
platform to improve care of patients with
cardiomyopathies using routine electronic health
records and standardised biobanking. Netherlands
Heart Journal, 27(9), 426–434. https://doi.org/10.10
07/s12471-019-1288-4
Sung, S.-F., Chen, K., Wu, D. P., Hung, L.-C., Su, Y.-H.,
& Hu, Y.-H. (2018). Applying natural language
processing techniques to develop a task-specific EMR
interface for timely stroke thrombolysis: A feasibility
study. International Journal of Medical Informatics,
112, 149–157. https://doi.org/10.1016/j.ijmedinf.20
18.02.005
Tomczak, K., Czerwińska, P., & Wiznerowicz, M. (2015).
Review<br>The Cancer Genome Atlas (TCGA): An
immeasurable source of knowledge. Contemporary
Oncology/Współczesna Onkologia, 2015(1), 68–77.
https://doi.org/10.5114/wo.2014.47136
Tukur, M., Saad, G., AlShagathrh, F. M., Househ, M., &
Agus, M. (2023). Telehealth interventions during
COVID-19 pandemic: A scoping review of
applications, challenges, privacy and security issues.
BMJ Health & Care Informatics, 30(1), Article 1.
https://doi.org/10.1136/bmjhci-2022-100676
Wu, S. T., Liu, H., Li, D., Tao, C., Musen, M. A., Chute, C.
G., & Shah, N. H. (2012). Unified Medical Language
System term occurrences in clinical notes: A large-scale
corpus analysis. Journal of the American Medical
Informatics Association, 19(e1), Article e1.
https://doi.org/10.1136/amiajnl-2011-000744
Yang, J., Jiang, Z., Benthin, F., Hanel, J., Fandrich, T., Joos,
R., Bauer, S., Kolatschek, S., Hreibi, A.,
Rugeramigabo, E. P., Jetter, M., Portalupi, S. L., Zopf,
M., Michler, P., Kück, S., & Ding, F. (2024). High-rate
intercity quantum key distribution with a
semiconductor single-photon source. Light: Science &
Applications,
13(1), 150. https://doi.org/10.1038/s413
77-024-01488-0
Yang, R., Tan, T. F., Lu, W., Thirunavukarasu, A. J., Ting,
D. S. W., & Liu, N. (2023). Large language models in
health care: Development, applications, and challenges.
Health Care Science, 2(4), Article 4.
https://doi.org/10.1002/hcs2.61
DATA 2025 - 14th International Conference on Data Science, Technology and Applications
472