Science 28Feb2020

(lily) #1

INSIGHTS | PERSPECTIVES


sciencemag.org SCIENCE

MEDICINE


Artificial intelligence in cancer therapy


Artificial intelligence can optimize cancer drug discovery, development, and administration


By Dean Ho


A

rtificial intelligence (AI) approaches
have the potential to affect several
facets of cancer therapy. These in-
clude drug discovery and devel-
opment and how these drugs are
clinically validated and ultimately
administered at the point of care, among
others. Currently, these processes are ex-
pensive and time-consuming. Moreover,
therapies often result in variable treatment
outcomes between patients. The conver-
gence of AI and cancer therapy has resulted
in multiple solutions to address these chal-
lenges. AI platforms ranging from machine
learning to neural networks can accelerate
drug discovery, harness biomarkers to ac-
curately match patients to clinical trials,
and truly personalize cancer therapy using
only a patient’s own data. These advances
are indicators that practice-changing can-
cer therapy empowered by AI may be on
the horizon.
AI is already making progress in acceler-
ating drug discovery and has successfully
predicted drug behavior by using genomic
and chemical data in lieu of large-scale
screening assays, which may accelerate
drug repurposing (that is, using drugs indi-
cated for one disease to treat other diseases)
( 1 ). Reinforcement learning, which uses re-
ward and punishment to train algorithms
to achieve a desired drug structure, success-
fully designed a new compound in just 21
days versus conventional timelines of ap-
proximately 1 year. Subsequently observed
pharmacokinetic properties suggested that
threshold drug exposure and efficacy could
be attained, supporting further evaluation
of the lead compound ( 2 ). In this study, the
generative tensorial reinforcement learning
(GENTRL) platform was trained by using a
dataset of chemical structures that target
the tyrosine kinase discoidin domain re-
ceptor 1 (DDR1), which is involved in the
progression of multiple cancers. Computer
simulations predicted lead compound and
receptor binding, aiming to minimize tar-
geting of other tyrosine kinases and en-
hance DDR1 targeting. Although additional
compound optimization is required, this is


an important step toward AI-accelerated
cancer drug discovery.
AI-enhanced drug discovery represents
one stage of a pipeline of processes needed
to optimize cancer therapy (see the figure).
During traditional drug development, ap-
proved and investigational compounds
are often codelivered in combination pre-
clinically and clinically to address multiple
drug targets, improving treatment efficacy.
Subsequent clinical dose escalation studies
identify the doses that achieve drug syn-
ergy, in which the drug combination has a
greater effect together than individually.
Unfortunately, issues such as off-target ef-
fects can preclude drug approval because
of unforeseen toxicity ( 3 ). Additionally,
well-designed compounds delivered at sub-
optimal doses may limit efficacy. Therefore,
optimized combination therapy design
simultaneously identifies the best drugs
for combination and doses that address
the right targets while minimizing toxic-
ity. Testing all possible drug combinations
at multiple doses for each drug is virtually
impossible. However, AI can overcome this
challenge by markedly reducing the number
of experiments needed to resolve drug and
dose parameters, optimizing combination
therapy development.
Achieving drug synergy is a key objective
of traditional approaches to design combina-
tion therapies to improve efficacy. However,
patient responses to combination therapy
are highly variable. Computational modeling
showed that effective combination therapy
can be realized without drug additivity or
synergy; a combination of drugs that act
independently with favorable efficacy may
improve treatment outcomes over synergy-
driven combinations ( 4 ). Modeling tumor
growth kinetics and drug sensitivity predic-
tions by using data from clinical trials that
recruited large cohorts of patients with dif-
ferent cancer types demonstrated that maxi-
mizing independent drug efficacy is a major
determinant of treatment response.
AI will play a vital role in designing drug
combinations without relying on synergy-
based modeling or predicted synergy be-
tween different drug targets and pathways.
This may markedly increase the pool of
drugs considered for treatment and identify
unexpected combinations that outperform
standards of care. When the dose of each
candidate drug is also considered, the pos-

sible drug and dose combinations are too
extensive for comprehensive validation.
However, AI can rapidly resolve large drug
and dose parameter spaces. For example,
the quadratic phenotypic optimization plat-
form (QPOP) uses quadratic relationships
represented by parabolas to visually corre-
late a set of inputs (e.g., drugs and doses)
with optimal outputs (e.g., preclinical tu-
mor reduction with minimal toxicity). This
correlation markedly reduces the number
of experiments and data needed to identify
the drugs and doses that optimize combina-
tion therapy design. Furthermore, QPOP is
agnostic to disease mechanisms, drug tar-
gets, and drug synergy.
The QPOP platform evaluated 14 chemo-
therapy drugs to treat multiple myeloma
(MM) with combination therapy ( 5 ). The
resulting drug combinations—such as
decitabine and mitomycin C, which were
unexpectedly and agonistically identified
by QPOP—markedly improved outcomes
in a MM mouse model compared with the
clinical standard-of-care drug combina-
tions ( 5 ). Importantly, neither decitabine
nor mitomycin c monotherapy mediated
efficacy alone, but their co-administration
optimally and synergistically reduced tu-
mor burden. Expanding QPOP to build
combinations of targeted therapies and
immunotherapies will be an important
next step toward treatment strategies be-
yond chemotherapy.
As oncology drug candidates move into
clinical validation, their regulatory ap-
proval rates have been reported to be as
low as 3.4% ( 6 ). Recent advances in trial
design have used biomarkers, such as ge-
nomic alterations, because of their poten-
tial as predictive indicators of treatment
response to stratify patient recruitment.
Including biomarkers in study recruitment
has improved patient outcomes compared
with traditional stratification information
such as pathology or responses to prior
treatments ( 3 , 7 ). Combining patient bio-
marker data and electronic health records
(EHRs) for AI analysis may further affect
trial outcomes ( 8 ). In the SYNERGY-AI
study (NCT03452774), virtual tumor
boards, which enable clinician teams to
provide remote treatment guidance and
diagnosis input, and EHRs pair oncol-
ogy patients with suitable clinical trials.
Multiple disease factors outlined by EHRs

The N.1 Institute for Health (N.1), Institute for Digital
Medicine (WisDM), Department of Biomedical Engineering,
and Department of Pharmacology, National University of
Singapore, Singapore. Email: [email protected]


982 28 FEBRUARY 2020 • VOL 367 ISSUE 6481


Published by AAAS
Free download pdf