Science 28Feb2020

(lily) #1
SCIENCE sciencemag.org

GRAPHIC: A. KITTERMAN/


SCIENCE


will be correlated with progression-free
survival and overall survival to assess the
effectiveness of the trial-matching plat-
form. Although many types of data may be
useful for stratification, AI will ultimately
need both population-scale and individu-
alized data to ensure that patients given
therapies, including those designed by AI,
have a high likelihood of responding.
AI will also play a pivotal role in how
cancer therapy is administered. Maximum
tolerated dosing eliminates drug-sensitive
tumor cells. However, drug-resistant cells
can eventually cause treatment failure.
Game theory is being explored to address
this challenge, with dose-reduction algo-
rithms competing against the tumor to
prevent drug-resistant cells (which have
high energy costs) from outnumbering

drug-sensitive cells ( 9 , 10 ). This is known
as adaptive therapy and may prolong treat-
ment efficacy by maintaining threshold
drug-sensitive cell populations in a tumor
to combat drug-resistant cell prolifera-
tion. Adaptive therapy was recently used
to modify paclitaxel dosing to treat mouse
models of breast cancer. Specifically, an
adaptive therapy algorithm (AT-1) itera-
tively reduced paclitaxel dosing after ob-
served tumor size reductions. AT-1 was
compared with a fixed-dosage algorithm
that halted drug dosing after observed
tumor reduction (AT-2) and high-dose
standard therapy ( 11 ). The AT-1 algorithm
improved tumor control and survival com-
pared with AT-2 and standard therapy,
demonstrating that game theory–driven
dosing can improve treatment outcomes
over established therapies at high doses.

Adaptive therapy was translated into a
pilot study for prostate cancer patients on
abiraterone hormone therapy ( 12 ). The adap-
tive therapy cohort received, on average, 47%
of the standard abiraterone dose, and three
of the patients received less than 25% of the
conventional dose. At the time of reporting,
1 of 11 adaptive therapy participants expe-
rienced tumor progression. This led to an
estimated median time to progression, us-
ing the biomarker prostate-specific antigen
(PSA) and radiographic imaging, of no less
than 27 months in this cohort, which was su-
perior to 11.1 months (PSA) and 16.5 months
(radiographic) for patients on continuous
abiraterone therapy ( 12 ). To maximize the
benefits of this platform, personalizing adap-
tive therapy by use of each patient’s response
to therapy versus population-based dose ad-

justment rules will likely be needed.
To further personalize patient-specific
combination therapy administration with
AI, CURATE.AI, a neural network–derived
platform, modulated multidrug dosing
using only a patient’s data by means of a
second-order algebraic algorithm that dy-
namically related dosing to optimal tumor
reduction and safety at any treatment time
point ( 13 , 14 ). In a patient treated with
enzalutamide hormone therapy and an in-
vestigational bromodomain and extrater-
minal domain (BET) inhibitor, data such as
physician-guided dose variations and corre-
sponding amounts of PSA were used to rec-
ommend a 50% reduction of the BET inhibi-
tor dose to increase efficacy. Subsequent
dynamic dosing of both drugs resulted in a
durable response and halted tumor progres-
sion, which was confirmed by computed

tomography (CT) imaging ( 13 ). This study
successfully used AI to modulate experi-
mental therapy dosing, demonstrating that
dosage guidance, without requiring big data
and complex genetic information, markedly
enhanced treatment efficacy compared with
fixed- and high-dose chemotherapy. Future
studies will need to evaluate whether this
platform can be implemented with addi-
tional classes of disease markers. Also, be-
cause of the individualized nature of this
platform, it will need to be further evalu-
ated in larger patient cohorts. As such, it is
being tested in a clinical trial that involves
a large pool of patients with hematological
malignancies (NCT03759093).
AI represents a gateway to the next fron-
tier of cancer therapy, reconciling extraor-
dinary amounts and different types of data
into actionable therapy. Among its barriers
to deployment are its siloed use in narrow
segments of the cancer therapy workflow.
For example, AI-optimized compounds
that are suboptimally combined with other
therapies or dosed incorrectly are unlikely
to substantially improve patient outcomes.
Overcoming this challenge in oncology will
require the seamless implementation of AI
across the spectrum of discovery, develop-
ment, and administration. Its potential
downstream applications include increas-
ing the resolution of personalized care by
tailoring bespoke regimens that integrate
multiple therapeutic strategies. For exam-
ple, AI-optimized radiation therapy dosing,
which maintained robust tumor size con-
trol, can potentially be combined with AI-
driven drug administration ( 15 ). Ultimately,
comprehensively adapting AI into clinical
oncology practice may improve drug acces-
sibility and reduce health-care costs. As AI
continues to be validated and a path toward
widespread practice is identified, its poten-
tial to redefine the clinical standards of can-
cer therapy is becoming evident. j
REFERENCES AND NOTES


  1. M. P. Menden et al., PLOS ONE 8 , e61318 (2013).

  2. A. Zhavoronkov et al., Nat. Biotechnol. 37 , 1038 (2019).

  3. A. Lin et al., Sci. Transl. Med. 11 , eaaw8412 (2019).

  4. A. C. Palmer, P. K. Sorger, Cell 171 , 1678 (2017).

  5. M. B. M. A. Rashid et al., Sci. Transl. Med. 10 , eaan0941
    (2018).

  6. C. H. Wong et al., Biostatistics 20 , 273 (2019).

  7. S. Harrer et al., Trends Pharmacol. Sci. 40 , 577 (2019).

  8. A. Rajkomar et al., NPJ Digit. Med. 1 , 18 (2018).

  9. J. Chmielecki et al., Sci. Transl. Med. 3 , 90ra59 (2011).

  10. C. Jedeszko et al., Sci. Transl. Med. 7 , 282ra250 (2015).

  11. P. M. Enriquez-Navas et al., Sci. Transl. Med. 8 , 327ra24
    (2016).

  12. J. Zhang et al., Nat. Commun. 8 , 1816 (2017).

  13. A. J. Pantuck et al., Adv. Ther. 1 , 1800104 (2018).

  14. A. Zarrinpar et al., Sci. Transl. Med. 8 , 333ra49 (2016).

  15. B. Lou et al., Lancet Digit. Health 1 , e136 (2019).
    ACKNOWLEDGMENTS
    D.H. holds pending patents on AI-based personalized
    medicine. D.H. acknowledges support from AI Singapore
    (NRF), the National Medical Research Council, and the
    Ministry of Education.
    10.1126/science.aaz3023


Tar g e t
identifcation

Combination
therapy design

Patient-therapy
matching

Dose

Time

Tar g e t

Discovery

Lead

Lead generation
and optimization

Preclinical
development

Clinical trials
phase I–III

Personalized
cancer therapy

Opportunities
Minimize of-target efects and toxicity
Enhance drug exposure

Challenges
Identifying optimal targets
Properly validating AI-designed drugs

Development
Opportunities
Optimize drug and dose selection
Match patients to therapies and trials

Challenges
Improving trial outcomes
Stratifcation with the right patient data

Administration
Opportunities
Sustained dose optimization
Overcoming resistance with
game theory
Challenges
More clinical validation needed
Use in more cancer types

28 FEBRUARY 2020 • VOL 367 ISSUE 6481 983

Improving multiple aspects of cancer therapy
Cancer therapy involves different stages, including drug discovery, development, and administration.
Artificial intelligence (AI) is poised to benefit each stage but is also confronted by challenges that, when
overcome, may lead to practice-changing cancer treatment.

Published by AAAS
Free download pdf