ArticlePDF Available

Enhancing Next‐Generation Sequencing‐Guided Cancer Care Through Cognitive Computing

Authors:

Abstract and Figures

Background: Using next-generation sequencing (NGS) to guide cancer therapy has created challenges in analyzing and reporting large volumes of genomic data to patients and caregivers. Specifically, providing current, accurate information on newly approved therapies and open clinical trials requires considerable manual curation performed mainly by human "molecular tumor boards" (MTBs). The purpose of this study was to determine the utility of cognitive computing as performed by Watson for Genomics (WfG) compared with a human MTB. Materials and methods: One thousand eighteen patient cases that previously underwent targeted exon sequencing at the University of North Carolina (UNC) and subsequent analysis by the UNCseq informatics pipeline and the UNC MTB between November 7, 2011, and May 12, 2015, were analyzed with WfG, a cognitive computing technology for genomic analysis. Results: Using a WfG-curated actionable gene list, we identified additional genomic events of potential significance (not discovered by traditional MTB curation) in 323 (32%) patients. The majority of these additional genomic events were considered actionable based upon their ability to qualify patients for biomarker-selected clinical trials. Indeed, the opening of a relevant clinical trial within 1 month prior to WfG analysis provided the rationale for identification of a new actionable event in nearly a quarter of the 323 patients. This automated analysis took <3 minutes per case. Conclusion: These results demonstrate that the interpretation and actionability of somatic NGS results are evolving too rapidly to rely solely on human curation. Molecular tumor boards empowered by cognitive computing could potentially improve patient care by providing a rapid, comprehensive approach for data analysis and consideration of up-to-date availability of clinical trials. Implications for practice: The results of this study demonstrate that the interpretation and actionability of somatic next-generation sequencing results are evolving too rapidly to rely solely on human curation. Molecular tumor boards empowered by cognitive computing can significantly improve patient care by providing a fast, cost-effective, and comprehensive approach for data analysis in the delivery of precision medicine. Patients and physicians who are considering enrollment in clinical trials may benefit from the support of such tools applied to genomic data.
Content may be subject to copyright.
Enhancing Next-Generation Sequencing-Guided Cancer Care Through
Cognitive Computing
NIRALI M. PATEL,
a,b,†
VANESSA V. MICHELINI,
f,†
JEFF M. SNELL,
a,c
SAIANAND BALU,
a
ALAN P. HOYLE,
a
JOEL S. PARKER,
a,c
MICHELE C. HAYWARD,
a
DAVID A. EBERHARD,
a,b
ASHLEY H. SALAZAR,
a
PATRICK MCNEILLIE,
g
JIA XU,
g
CLAUDIA S. HUETTNER,
g
TAKAHIKO KOYAMA,
h
FILIPPO UTRO,
h
KAHN RHRISSORRAKRAI,
h
RAQUEL NOREL,
h
ERHAN BILAL,
h
AJAY ROYYURU,
h
LAXMI PARIDA,
h
H. SHELTON EARP,
a,d
JUNEKO E. GRILLEY-OLSON,
a,d
D. NEIL HAYES,
a,d
STEPHEN J. HARVEY,
i
NORMAN E. SHARPLESS,
a,c,d
WILLIAM Y. KIM
a,c,d,e
a
Lineberger Comprehensive Cancer Center,
b
Department of Pathology and Laboratory Medicine,
c
Department of Genetics,
d
Department of
Medicine, and
e
Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA;
f
IBM Watson Health,
Boca Raton, Florida, USA;
g
IBM Watson Health, Cambridge, Massachusetts, USA;
h
IBM Research, Yorktown Heights, New York, USA;
i
IBM
Watson Health, Herndon, Virginia, USA
Contributed equally
Disclosures of potential conflicts of interest may be found at the end of this article.
Key Words. Genomics High-throughput nucleotide sequencing Artificial intelligence Precision medicine
ABSTRACT
Background. Using next-generation sequencing (NGS) to guide
cancer therapy has created challenges in analyzing and report-
ing large volumes of genomic data to patients and caregivers.
Specifically, providing current, accurate information on newly
approved therapies and open clinical trials requires consider-
able manual curation performed mainly by human “molecular
tumor boards” (MTBs). The purpose of this study was to deter-
mine the utility of cognitive computing as performed by Wat-
son for Genomics (WfG) compared with a human MTB.
Materials and Methods. One thousand eighteen patient cases
that previously underwent targeted exon sequencing at the
University of North Carolina (UNC) and subsequent analysis
by the UNCseq informatics pipeline and the UNC MTB
between November 7, 2011, and May 12, 2015, were
analyzed with WfG, a cognitive computing technology for
genomic analysis.
Results. UsingaWfG-curatedactionablegenelist,weidentied
additional genomic events of potential significance (not discov-
ered by traditional MTB curation) in 323 (32%) patients. The
majority of these additional genomic events were considered
actionable based upon their ability to qualify patients for
biomarker-selected clinical trials. Indeed, the opening of a rele-
vant clinical trial within 1 month prior to WfG analysis provided
the rationale for identification of a new actionable event in
nearly a quarter of the 323 patients. This automated analysis
took <3 minutes per case.
Conclusion. These results demonstrate that the interpretation
and actionability of somatic NGS results are evolving too rapidly
to rely solely on human curation. Molecular tumor boards
empowered by cognitive computing could potentially improve
patient care by providing a rapid, comprehensive approach for
data analysis and consideration of up-to-date availability of
clinical tr ials. The Oncologist 2017;22:1–7
Implications for Practice: The results of this study demonstrate that the interpretation and actionability of somatic next-generation
sequencing results are evolving too rapidly to rely solely on human curation. Molecular tumor boards empowered by cognitive
computing can significantly improve patient care by providing a fast, cost-effective, and comprehensive approach for data analysis
in the delivery of precision medicine. Patients and physicians who are considering enrollment in clinical trials may benefit from the
support of such tools applied to genomic data.
INTRODUCTION
Next-generation sequencing (NGS) has emerged as an afford-
able and reproducible means to query patients’ tumors for
somatic genetic anomalies [1, 2]. The optimal utilization of NGS
is fundamental to the promise of “precision medicine,” yet the
results of even targeted-capture NGS are highly complex,
returning a variety of somatic events in hundreds of analyzed
genes. The majority of such events have no known relevance
to the treatment of patients with cancer, and even for
Correspondence: William Y. Kim, M.D., or Norman E. Sharpless, M.D., Lineberger Comprehensive Cancer Center, University of North Carolina, CB#
7295, Chapel Hill, North Carolina 27599-7295, USA. Telephone: 919-966-4765; e-mail: william_kim@med.unc.edu or nes@med.unc.edu Received
April 18, 2017; accepted for publication October 6, 2017. http://dx.doi.org/10.1634/theoncologist.2017-0170
The Oncologist 2017;22:1–7 www.TheOncologist.com
O
cAlphaMed Press 2017
Cancer Diagnostics and Molecular Pathology
Published Ahead of Print on November 20, 2017 as 10.1634/theoncologist.2017-0170.
by guest on November 16, 2018http://theoncologist.alphamedpress.org/Downloaded from
well-understood driver events (e.g., BRAF V600E), the combina-
torial significance of other mutations is poorly understood.
Tumors may also possess a large degree of intratumor hetero-
geneity [3], adding further complexity to the analysis.
In order to handle somatic NGS data, many institutions
have created a “molecular tumor board” (MTB) made up of
physicians and biomedical scientists to analyze the results of
NGS and make recommendations. MTBs in turn rely on an
“actionable gene list” of specific genetic events that should be
considered of clinical significance. Mutations on this list can
guide therapy (e.g., BRAF mutations in melanoma), provide
prognostic information (e.g., FLT3 mutations in acute myeloge-
nous leukemia), or be of diagnostic utility (e.g., EWS-FLI translo-
cations in a poorly differentiated pediatric sarcoma). At
present, the state of our clinical knowledge regarding the
actionability of genetic events evolves and grows sporadically,
largely through the reporting of new clinical results in a variety
of scientific venues. As thousands of new cancer-relevant publi-
cations are produced daily, staying abreast of new clinical infor-
mation in oncology is extremely challenging and differs from
institution to institution.
In 2015, there were more than 2.5 million scholarly articles
published, with more than 150,000 directly related to cancer, a
volume of literature that is growing at an estimated rate of
6.7% per year over the past decade [3]. At the time of submis-
sion of this article, a total number of 61 targeted inhibitors are
approved for the treatment of a few conditions. There are
more than 650 targeted therapies in development, with thou-
sands of clinical trials in progress. In order to be used by current
computer systems, relevant data from these articles and clinical
trials, which are written in a free-text format (unstructured
data), need to be extracted, cleaned, translated to canonical
forms, validated, and formatted in structured tables and data-
bases. In many cases, this process is done manually by subject
matter experts who must read the free text and transpose the
information into a structured format. However, with such rapid
growth of new data, this is both expensive and not a practical,
scalable approach.
MATERIALS AND METHODS
Targeted Exon Sequencing
Targeted exon sequencing was conducted after institutional
review board (IRB) approval through the UNCseq (University of
North Carolina, Chapel Hill, NC, http://www2.cscc.unc.edu/
unchreg/UNCseq) pipeline, as previously described [4, 5]. In
brief, sequencing data are routed through an automated pipe-
line. This workflow uses paired tumor and normal libraries to
detect somatic mutations, large and small indels, structural var-
iants, and copy number aberrations. Raw sequences are aligned
using the Burrows-Wheeler Aligner (BWA)-mem algorithm and
refined using our Assembly Based ReAlignment (ABRA [6]) pro-
cess. Our UNCeqR [7] algorithm, combined with Strelka [8],
then provides sensitive detection of somatic variants.
UNCseq Actionability
Genes were defined as “actionable” by the University of North
Carolina (UNC) MTB if they met criteria that placed them in
Tier 1 (variant targeted by commercially available drug that is
approved to treat this specific genetic variation), Tier 2A
(variant potentially treatable by commercially available tar-
geted drug, but the drug is not indicated for this use), or Tier
2B (variant is potentially treatable by targeted drug that is in
clinical trials) [1].
All variants of unknown significance (VUS) are evaluated by
the UNC MTB and, based upon characteristics of the VUS and a
search of the published literature, are considered for return
into the medical record. Not all VUSs are returned to the medi-
cal record.
Watson for Genomics
Upon sequence completion by UNCseq, the following informa-
tion was uploaded to Watson for Genomics (WfG): (a) tumor
type, (b) a list of variants as a variant calling file (.vcf), and (c)
gene level copy number alterations as a log 2 ratio of tumor to
UNCseq
MTB
UNCseq
informatics pipeline
WfG
UNCseq MTB
actionable genes
Variants
Copy number alterations
WfG
actionable genes
Figure 1. Study outline. Sequencing data from 1,018 cases were run
through the UNCseq informatics pipeline to generate lists of variants
and copy number alterations. Genomic profiles for each patient
were reviewed at the UNCseq MTB and, based on the genomic
alteration and its presence on the list of actionable genes previously
determined by the UNCseq Clinical Committee for Genomic
Research, were deemed actionable. For each patient, WfG was pro-
vided information on the type of cancer and the full list of variants
and copy number alterations that had been detected by the UNCseq
informatics pipeline to derive its list of actionable genomic events.
Abbreviations: MTB, molecular tumor board; UNC, University of
North Carolina; WfG, Watson for Genomics.
2Enhancing Cancer Care Through Cognitive Computing
O
cAlphaMed Press 2017
Published Ahead of Print on November 20, 2017 as 10.1634/theoncologist.2017-0170.
by guest on November 16, 2018http://theoncologist.alphamedpress.org/Downloaded from
normal. After the above information was uploaded, the follow-
ing steps were executed by WfG for each gene with a variant or
copy number alteration (supplemental online Fig. 1):
1. Molecular Profile Analysis (MPA).Evidence from functional
studies and protein structure, combined with programming
logic, was used to classify variants into five categories: patho-
genic, likely pathogenic, benign, likely benign, and VUS. Benign
and likely benign mutations were removed from the report.
2. Actionable alterations analysis.Once the alterations were
categorized by the MPA, WfG assigned a gene as actionable
if (a) the variant was pathogenic or likely pathogenic, (b)
the variant was directly targetable or part of a pathway that
was targetable based on evidence from the literature, and
(c) a U.S. Food and Drug Administration-approved or investi-
gational target therapy was available.
3. Drug analysis.In this step, potential therapeutic options
were associated with actionable mutations. Note that only
clinical trials actively recruiting were considered during this
step. Analysis of resistance was also performed during this
step. WfG uses levels of evidence (Table 2) for the gene, var-
iant, cancer type, and drug association.
4. Report.A report was generated by WfG showing the var-
iants (pathogenic, likely pathogenic, and VUS) alongside
potential targeted drugs. An example of the user interface
is shown in supplemental online Figure 1.
The supplemental online Methods provides further details
on WfG natural language processing (NLP) techniques and liter-
ature extraction. Supplemental online Figure 1 outlines the
workflow for Watson for Genomics Analysis, representing the
four steps described above.
RESULTS
Several cancer centers have started using cognitive computing
systems [9], yet few data exist on the validity of these
approaches or their promise to deliver personalized medicine
to patients. Cognitive computing is a form of artificial intelli-
gence (AI) that relies on machine learning, NLP, and other data
analysis technologies to understand and draw patterns from
massive volumes of disparate data. Cognitive computing
approaches such as WfG rely on computerized models to simu-
late aspects of human thought. In particular, using state-of-the-
art machine learning, NLP, and cognitive insights, WfG is well
suited to ingest, organize, aggregate, and extract relevant
insights from large volumes of rapidly emerging clinical data.
Although cognitive systems can ingest new information in real
time, WfG uses an offline process that consists of extraction,
validation, and testing before the insights are incorporated into
real-time analysis (supplemental online Fig. 1). Importantly,
WfG can learn new information and analyze data at a rate that
far exceeds manual curation and analysis [10].
In order to compare the effectiveness of the WfG cognitive
computing engine with human-only MTBs at identifying treat-
ment options for patients, we retrospectively examined 1,018
consecutive cases from UNC that had undergone targeted DNA
sequencing (supplemental online Table 1) of matched tumor
and normal tissue and subsequent analysis by the UNCseq
informatics pipeline and the UNC MTB between November 7,
2011, and May 12, 2015 (Fig. 1). Baseline genomic characteris-
tics of the 1,018 patients demonstrated that the relative muta-
tional burden of different tumor types appeared to be in
keeping with the relative mutational burden previously
reported across TCGA tumors (e.g., melanoma, lung, and blad-
der cancers had the highest mutational burden; Fig. 2A and
supplemental online Table 2) [11]. A mean of 37% of protein-
altering mutations was considered actionable as defined by the
UNC MTB (Fig. 2B).
We next asked WfG to perform an independent analysis of
these 1,018 patients (Fig. 1). All 1,018 cases were analyzed by
WfG in November 2015. For each patient, WfG was provided
information on the type of cancer and the full list of variants
Number of protein-alterin
g
mutations
Sarcoma
Other
Thyroid
Hematologic
Upper GI
Kidney
Brain
Ovarian/peritoneal
Cervical
Breast
GI
Lymphoma
Head and neck
Uterus
Bladder
Lung
Melanoma
Number of mutations
0 5 10 15
Actionable
Total mutations
AB
Sarcoma
Other
Thyroid
Hematologic
Upper GI
Kidney
Brain
Ovarian/peritoneal
Cervical
Breast
GI
Lymphoma
Head and neck
Uterus
Bladder
Lung
Melanoma
0 102030405060
Figure 2. Mutational and actionable mutation burden by tumor type. (A): Median (dark line), interquartile range (box), and range
(whiskers) of protein-altering mutations by cancer type. (B): Number of actionable mutations (blue bar) as determined by the University
of North Carolina Molecular Tumor Board by cancer type.
Abbreviation: GI, gastrointestinal.
Patel, Michelini, Snell et al. 3
www.TheOncologist.com
O
cAlphaMed Press AlphaMed
Published Ahead of Print on November 20, 2017 as 10.1634/theoncologist.2017-0170.
by guest on November 16, 2018http://theoncologist.alphamedpress.org/Downloaded from
Additional mutations
Figure 4. Additional actionable mutations identified by WfG and not by the University of North Carolina Molecular Tumor Board (MTB),
categorized by tumor type. Tumor types are plotted on the y-axis and number of mutations are plotted on the x-axis. The solid circles rep-
resent the mean number of mutations identified by WfG and not by the MTB for each subtype, and the whiskers show the minimum and
maximum.
Abbreviations: GI, gastrointestinal; WfG,Watson for Genomics.
UNCseq pts
(1,018)
UNCseq pts
Actionable gene
(703)
UNCseq pts
No actionable gene
(315)
WfG
Actionable gene
(231)
WfG
Actionable gene
(96)
WfG
Actionable gene
(327)
WfG-identified actionable
gene approved by CCGR
(323)
Potential to change therapy (47)
No evidence of disease (145)
Lost to follow-up (29)
Withdrew from study (4)
Deceased (98)
AB
CDE
Figure 3. Sankey diagram of the flow of the UNCseq molecular tumor board (MTB) and WfG comparison. Of the 1,018 patients previously ana-
lyzed by the University of North Carolina (UNC) MTB, 703 were determined to have alterations in genes that met the UNC MTB definition of
actionability (A) and 315 did not (B).The WfG analysis suggested that an additional eight genes not previously defined as actionable should be
added to the actionable gene list. (C): Mutations in these eight genes were found in 231 and 96 patients out of the 703 and 315 patients with
actionable mutations and no actionable mutations, respectively. (D): Of the eight newly identified WfG genes, seven passed the criteria for
actionability as determined by the UNC CCGR. Mutations in at least one of these seven genes were found in 323 patients. (E): Re-examination
of these 323 patients revealed that while 47 had potential to change therapy, the majority of patients did not have the potential to change
therapy for several reasons (no evidence of disease, n5145; lost to follow-up, n529; withdrew from study, n54; and deceased, n598).
Abbreviations: CCGR, Clinical Committee for Genomic Research; pts, patients; WfG, Watson for Genomics.
4Enhancing Cancer Care Through Cognitive Computing
O
cAlphaMed Press 2017
Published Ahead of Print on November 20, 2017 as 10.1634/theoncologist.2017-0170.
by guest on November 16, 2018http://theoncologist.alphamedpress.org/Downloaded from
and copy number alterations that had been detected by the
UNCseq informatics pipeline (for details, see the Materials
and Methods section). The WfG analysis took approximately 3
minutes per case in this study. Of these 1,018 cases, the
human-only MTB had previously requested validation of
actionable variants in 703 of the patients (Fig. 3).These results
were reported to the treating physician and included in the
patient record in accordance with the IRB-approved UNCseq
protocol (LCCC1108/IRB #11–1115). In its independent analy-
sis of these 1,018 patients, WfG also categorized as actionable
the entirety of the human-only MTB-defined actionable var-
iants in all 1,018 cases. This result shows that WfG is able to
identify all reportable genetic events found by an MTB using
standard practices, including a human-curated actionable
gene list.
In addition to identifying reportable events already con-
veyed by the UNC MTB, WfG detected actionable variants in
323 (32%) of cases across a spectrum of tumor types (Fig. 4)
that were not reported by the UNC MTB (Fig. 4 and supplemen-
tal online Table 3). These events reflected eight genes that
were deemed actionable by WfG but not the human MTB at
the time of the reanalysis (Table 1). Importantly, even at the
time of the WfG analysis, these eight genes were still not on
the UNC MTB list of actionable genes, suggesting that our dis-
parate results were not merely reflecting the retrospective
nature of this study. Seven of the eight genes met the UNCseq
program’s own definition of actionability (e.g., being part of
entry criteria for a biomarker selected clinical trial; see the
Materials and Methods section), but were not identified by the
human-curated actionable gene list. These seven genes were
subsequently approved by the UNC Clinical Committee for
Genomic Research (CCGR). This left 323 patients with WfG-
identified actionable mutations that met the UNCseq definition
of actionability. The majority of newly actionable events discov-
ered by WfG were based on recent publications or newly
opened clinical trials for patients harboring inactivating events
of ARID1A,FBXW7,andATR (with or without concomitant
mutations of ATM;Table1).Ofthe323patientswithnewly
identified events, 283 (88%) patients were made potentially eli-
gible for enrollment in a biomarker-selected clinical trial that
had not been identified by the MTB. In particular, several of the
relevant clinical trials had opened and begun enrollment within
weeks of the WfG analysis (e.g., NCT02576444 for AIRD1A
mutant cancers). These observations suggest that staying
abreast of enrolling clinical trials is a particular challenge for
human-curated MTBs, and this problem is likely to worsen
as larger numbers of genotype-driven studies are opened
nationally.
As the WfG analysis was performed retrospectively on
selected patients, new findings were not relevant to the major-
ity of patients analyzed. In most cases, patients were not candi-
dates for further therapy for a variety of reasons (e.g.,
deceased, n598; no evidence of disease, n5145). Despite
the historical nature of the analysis, the MTB determined that
the results of the WfG analysis were of value in 47 patients
with active disease potentially needing further therapy. In
these cases, the NGS results were confirmed by a standard-of-
care, Clinical Laboratory Improvement Amendments-approved
assay in accord with the UNCseq guidelines, and then included
in the patient’s medical records and reported to the treating
physicians. To our knowledge, no patient with a WfG-identified
potentialtherapywentontobeenrolledinaclinicaltrial.
Table 1. Description of actionable findings in 283 patients identified by WfG missing in MTB recommendations
Gene n
CCGR
before
CCGR
after
WfG-identified
therapy
Clinical
trial Ph.
Rec.
agent
Ref.
(PMID)
APC 57 Not actionable Beta-catenin inhibitor Y II PRI-724 26396911
ARID1A 176 Not actionable PARPi Y II AZD2881 26069190
ATR 82 Not actionable PARPi Y I/Ib AZD6378 26517239
BRIP1 9 Not actionable PARPi Y II BMN 673 23881923
CDKN2B 13 Not Not CDK4/6i Y III Palbociclib 20354191
FBXW7 45 Not actionable mTORi N N/A N/A 24360397
MITF 3 Not actionable BRAFi N N/A N/A 24265153
RAD50 20 Not actionable Irinotecan 1Chk1/2i N N/A N/A 24934408
Abbreviations: CCGR, UNC Clinical Committee for Genomic Research; MTB, molecular tumor board; N, no; N/A, not applicable; Ph., clinical trial
phase; PMID, PubMed identifier; Rec. agent, recommended agent; Ref., reference; WfG, Watson for Genomics; Y, yes.
Table 2. Levels of evidence used by Watson for Genomics
Level Description
1 FDA-approved drug in this cancer type and biomarker
2A Standard of care biomarker predictive of response to an FDA-approved drug in this indication
2B Standard of care biomarker predictive of response to an FDA-approved drug in a different indication
3A Compelling clinical evidence supports the biomarker as being predictive of response to a drug in this indication
3B Compelling clinical evidence supports the biomarker as being predictive of response to a drug in a different indication
4 Compelling biological evidence supports the biomarker as being predictive of response to a drug
R1 Standard of care biomarker predictive of resistance to an FDA-approved drug in this indication
Abbreviation: FDA, U.S. Food and Drug Administration.
Patel, Michelini, Snell et al. 5
www.TheOncologist.com
O
cAlphaMed Press AlphaMed
Published Ahead of Print on November 20, 2017 as 10.1634/theoncologist.2017-0170.
by guest on November 16, 2018http://theoncologist.alphamedpress.org/Downloaded from
DISCUSSION
In this work, we employed Watson for Genomics, a cognitive
computing approach with sophisticated NLP, to extract relevant
information from unstructured data and to identify actionable
insights in patients enrolled in a clinical sequencing program.
Through ingestion of thousands of cancer-relevant publications
and analysis of ongoing clinical trials, WfG was able to identify
the same actionable events reported by the human-only MTB.
In addition, WfG identified new actionable events in more than
a quarter of the patients in this study. The majority of these
genetic events met criteria for enrollment in clinical trials, but a
minority also potentially predicted response to off-label therapy
and/or provided clinically meaningful prognostic information.
One reasonable response to this analysis is that a human
MTB can be infrequently informed by a cognitive computing
approach. For example, one might infer that a WfG analysis
could be run once on an institution’s actionable gene list and
then further human-only use would be adequate. This view,
however, misunderstands the fact that knowledge in this field is
rapidly evolving, with new publications and clinical trials chang-
ing the pathologic significance of a given genetic event on a near
daily basis. Therefore, the actionable gene list is never “finished,”
but instead requires real-time updating, which is a task to which
humans are poorly suited. Additionally, the evolving nature of a
given gene’s clinical utility provides an additional problem: a
given patient may not have actionable events at the time of ini-
tial diagnosis, but certain mutations may later become action-
able as research in the biomedical space progresses and
additional clinical trials become available. Therefore, reanalysis
of a tumor’s somatic mutations would seem prudent using
recent publication and trials information, but frequent reanalysis
of multiple patients would further increase the burden on over-
stretched MTBs. Therefore, the ability of a cognitive tool like
WfG increases the scalability of somatic tumor sequencing and
subsequent analysis of mutations for precision medicine.
Cognitive computational systems have shown promise in
other areas of medicine. For example, they have been success-
fully used to augment physicians’ ability to detect spinal lesions
in patient magnetic resonance images [12]. Medical cognitive
applications have also been found to be efficient for surgical
phase recognition and image processing for tumor progression
mappings [13]. In the arena of drug repurposing and drug target
identification, pilot studies have suggested that cognitive com-
puting can accelerate identification of novel drug targets [14].
Despite the power of cognitive computing approaches in these
other areas, however, to date their penetration into genomic
analysis has been limited. One reason for this has been frag-
mentation of medical data (i.e., the storage of data that cannot
be shared in multiple different formats across disparate plat-
forms); therefore, the large datasets required to train cognitive
computing approaches have not been available. This problem is
being directly addressed with several laudable data aggregation
efforts in both public and private settings (e.g., the American
Society of Clinical Oncology’s CancerLinQ and the American
Association for Cancer Research’s Project Genomics Evidence
Neoplasia Information Exchange). In this work, we were able to
identify a large amount of data (from http://www.ncbi.nlm.nih.
gov/pubmed and https://clinicaltrials.gov/, etc.) for an analysis
well suited to cognitive computing providing a highly useful
function for care of patients with cancer.
CONCLUSION
Although it is clear that cognitive computing will not substitute
for the ability of trained physicians and biomedical scientists to
interact with patients and to interpret clinical data, we have
reached a juncture at which many of the tasks related to cancer
care require a real-time analysis of large, continuously main-
tained datasets. Humans are poorly suited to tasks such as a
comprehensive analysis of the enormous amount of new clini-
cal information generated on a daily basis, and the present
study shows that a cognitive computing approach such as WfG
can enhance care for a significant fraction of patients.
Our study suggests that cognitive computing can expand
the treatment options for patients with cancer. However, the
majority of the 323 patients that WfG identified as having
actionable alterations were reclassified because an alteration in
the gene allowed them to be considered for enrollment in a
biomarker-selected clinical trial. We realize that these genes
have yet to demonstrate their capability as predictive bio-
markers of response to the proposed therapy and that while
patients may value the comprehensive nature of WfG, one
downside of the exhaustive nature of WfG may be the presen-
tation of too many options. Moreover, we recognize that the
mere identification of actionable and targetable genomic
events does not necessarily translate into patient benefit [15].
Results of the SHIVA trial question whether off-label use of
molecularly targeted agents does benefit patients [16], and this
topic remains debated in the medical literature [15, 17]. None-
theless, we remain in the early days of NGS-guided cancer care
and believe that the road to improved patient outcomes likely
lies in an iterative process that needs to be instituted between
MTB-recommended therapies and patient outcomes as well as
the availability of potent and effective therapies [1].
ACKNOWLEDGMENTS
We thank the members of the UNCseq team and the Line-
berger Bioinformatics Core for expertise and data analysis and
Arrow Genomics, LLC, for help with manuscript preparation.
Partial results of this study have previously been presented as
part of the CBS 60 Minutes segment on artificial intelligence.
This work was supported by the NC University Cancer Research
Fund and grant funding from NIH U10-CA181009 (D.N.H.) and
NIH R01-CA202053 (W.Y.K.).
AUTHOR CONTRIBUTIONS
Conception/design: Vanessa V. Michelini, David A. Eberhard, Patrick McNeillie,
Filippo Utro, Raquel Norel, Ajay Royyuru, H. Shelton Earp, Juneko E. Grilley-
Olson, D. Neil Hayes, Stephen J. Harvey, Norman E. Sharpless, William Y. Kim
Provision of study material or patients: Juneko E. Grilley-Olson, D. Neil Hayes,
Norman E. Sharpless, William Y. Kim
Collection and/or assembly of data: Nirali Patel, Vanessa V. Michelini, Saianand
Balu, Alan P. Hoyle, Joel S. Parker, Michele C. Hayward, David A. Eberhard,
Ashley H. Salazar, Patrick McNeillie, Raquel Norel, Erhan Bilal, D. Neil Hayes,
William Y. Kim
Data analysis and interpretation: Nirali Patel, Vanessa V. Michelini, David A.
Eberhard, Patrick McNeillie, Jia Xu, Claudia S. Huettner, Takahiko Koyama,
Filippo Utro, Kahn Rhrissorrakrai, Laxmi Parida, D. Neil Hayes,William Y. Kim
Manuscript writing: Nirali Patel, Vanessa V. Michelini, Jeff M. Snell, Saianand
Balu, Alan P. Hoyle, Joel S. Parker, Michele C. Hayward, David A. Eberhard,
Ashley H. Salazar, Patrick McNeillie, Jia Xu, Claudia S. Huettner, Takahiko
Koyama, Filippo Utro, Kahn Rhrissorrakrai, Raquel Norel, Erhan Bilal, Ajay
Royyuru, Laxmi Parida, H. Shelton Earp, Juneko E. Grilley-Olson, D. Neil Hayes,
StephenJ. Harvey, Norman E. Sharpless, William Y. Kim
Final approval of manuscript: Nirali Patel, Vanessa V. Michelini, Jeff M. Snell,
Saianand Balu, Alan P. Hoyle, Joel S. Parker, Michele C. Hayward, David A.
Eberhard, Ashley H. Salazar, Patrick McNeillie, Jia Xu, Claudia S. Huettner,
6Enhancing Cancer Care Through Cognitive Computing
O
cAlphaMed Press 2017
Published Ahead of Print on November 20, 2017 as 10.1634/theoncologist.2017-0170.
by guest on November 16, 2018http://theoncologist.alphamedpress.org/Downloaded from
Takahiko Koyama, Filippo Utro, Kahn Rhrissorrakrai, Raquel Norel, Erhan Bilal,
Ajay Royyuru, Laxmi Parida, H. Shelton Earp, Juneko E. Grilley-Olson, D. Neil
Hayes,Stephen J. Harvey, Norman E. Sharpless,William Y. Kim
DISCLOSURES
Vanessa V. Michelini:IBM(E,OI);Patrick McNeillie: IBM (E, OI); Jia Xu:
IBM (E, OI); Claudia S. Huettner: IBM (E, OI); Takahiko Koyama: IBM
(E, OI); Filippo Utro:IBM(E,OI);Kahn Rhrissorrakrai:IBM(E,OI);
Raquel Norel: IBM (E, OI); Erhan Bilal: IBM (E, OI); Ajay Royyuru:IBM
(E, OI); Laxmi Parida: IBM (E, OI); H. Shelton Earp: FORTHCOMING;
Stephen J. Harvey: IBM (E, OI); Norman E. Sharpless:G1
Therapeutics, Unity Biotechnology, HealthSpan Diagnostics (C/A, IP,
SAB, OI), Pfizer (H); William Y. Kim: IBM (C/A). The other authors
indicated no financial relationships.
(C/A)Consulting/advisory relationship;(RF) Researchfunding; (E) Employment; (ET) Expert
testimony; (H) Honoraria received; (OI) Ownership interests; (IP)Intellectualproperty rights/
inventor/patentholder; (SAB)Scientific advisory board
REFERENCES
1. Hayes DN, Kim WY. The next steps in next-gen
sequencing of cancer genomes. J Clin Invest 2015;
125:462–468.
2. Garraway LA. Genomics-driven oncology:
Framework for an emerging paradigm. J Clin Oncol
2013;31:1806–1814.
3. National Science Board, National Science Foun-
dation. Science and Engineering Indicators 2014.
Available at http://www.nsf.gov/statistics/seind14/.
Accessed September 10, 2016.
4. JeckWR,ParkerJ,CarsonCCetal.Targetednext
generation sequencing identifies clinically actionable
mutations in patients with melanoma. Pigment Cell
Melanoma Res 2014;27:653–663.
5. Zhao X, Wang A, Walter V et al. Combined tar-
geted DNA sequencing in non-small cell lung cancer
(NSCLC) using UNCseq and NGScopy, and RNA
sequencing using UNCqeR for the detection of
genetic aberrations in NSCLC. PLoS ONE 2015;10:
e0129280.
6. Mose LE, Wilkerson MD, Hayes DN et al.
ABRA: Improved coding indel detection via
assembly-based realignment. Bioinformatics
2014;30:2813–2815.
7. Wilkerson MD, Cabanski CR, Sun W et al. Inte-
grated RNA and DNA sequencing improves mutation
detection in low purity tumors. Nucleic Acids Res
2014;42:e107.
8. Saunders CT, Wong WS, Swamy S et al. Strelka:
Accurate somatic small-variant calling from
sequenced tumor-normal sample pairs. Bioinfor-
matics 2012;28:1811–1817.
9. American Association for Cancer Research.
News in Brief: Oncologists partner with Watson on
genomics. Cancer Discov 2015;5:788.
10. Wrzeszczynski KO, Frank MO, Koyama T et al.
Comparing sequencing assays and human-machine
analyses in actionable genomics for glioblastoma.
Neurol Genet 2017;3:e164.
11. Kandoth C, McLellan MD, Vandin F et al. Muta-
tional landscape and significance across 12 major
cancer types. Nature 2013;502:333–339.
12. Ogiela L, Tadeusiewicz R, Ogiela MR. Cognitive
techniques in medical information systems. Comput
Biol Med 2008;38:501–507.
13. Philipp P, Maleshkova M, Katic D et al. Toward
cognitive pipelines of medical assistance algorithms.
Int J Comput Assist Radiol Surg 2016;11:1743–1753.
14. Chen Y, Elenee Argentinis JD, Weber G. IBM
Watson: How cognitive computing can be applied to
big data challenges in life sciences research. Clin
Ther 2016;38:688–701.
15. West HJ. No solid evidence, only hollow argu-
ment for universal tumor sequencing: Show me the
data. JAMA Oncol 2016;2:717–718.
16. Le Tourneau C, Delord JP, Gonc¸alves A et al.
Molecularly targeted therapy based on tumour
molecular profiling versus conventional therapy for
advanced cancer (SHIVA): A multicentre, open-label,
proof-of-concept, randomised, controlled phase 2
trial. Lancet Oncol 2015;16:1324–1334.
17. Subbiah V, Kurzrock R. Universal genomic test-
ing needed to win the war against cancer: Genomics
IS the diagnosis. JAMA Oncol 2016;2:719–720.
See http://www.TheOncologist.com for supplemental material available online.
Patel, Michelini, Snell et al. 7
www.TheOncologist.com
O
cAlphaMed Press AlphaMed
Published Ahead of Print on November 20, 2017 as 10.1634/theoncologist.2017-0170.
by guest on November 16, 2018http://theoncologist.alphamedpress.org/Downloaded from

Supplementary resource (1)

... En las últimas décadas, se ha producido un avance significativo en el campo de la oncología gracias a la medicina de precisión y el uso de la inteligencia artificial (7). Esta se basa en el análisis detallado de las características moleculares de los tumores, permitiendo así un enfoque terapéutico más personalizado (8). Por otro lado, la inteligencia artificial ha demostrado su capacidad para analizar grandes volúmenes de datos y generar modelos predictivos que pueden ayudar en la toma de decisiones clínicas (4). ...
... La información obtenida a través de estas técnicas permite identificar alteraciones genéticas específicas en el ADN tumoral, lo que a su vez ayuda a seleccionar tratamientos dirigidos que atacan estas alteraciones específicas. Además, la medicina de precisión también permite predecir la respuesta al tratamiento y evaluar el riesgo de recurrencia del cáncer en cada paciente individual (7,8). ...
... Al utilizar la información genómica y molecular de los tumores, la medicina de precisión permite una personalización del tratamiento que maximiza la eficacia y minimiza los efectos secundarios. La inteligencia artificial complementa este enfoque al proporcionar herramientas de análisis y predicción que ayudan a los médicos a tomar decisiones más precisas y basadas en evidencia (4,7,8). ...
Article
Introducción: La inteligencia artificial (IA) ha revolucionado la medicina y la oncología al mejorar el diagnóstico, tratamiento y cirugía. La IA analiza grandes volúmenes de datos, identifica patrones y mejora la precisión clínica. En oncología, la IA y la medicina de precisión permiten un enfoque terapéutico personalizado. En imagenología médica, la IA mejora el diagnóstico de enfermedades como el cáncer de pulmón. Objetivo: Analizar la implementación y utilidad de la inteligencia artificial en las ciencias de la salud. Metodología: Se realizó un estudio no experimental, descriptivo, tipo revisión bibliográfica. se buscó información en PubMed, ScienceDirect y Scielo utilizando operadores boléanos sacados de DeCS y MESH. Desarrollo: La IA en la salud utiliza algoritmos de aprendizaje automático y asistentes virtuales para brindar atención médica accesible. En cirugía, la IA complementa la robótica quirúrgica y mejora la precisión. Aunque presenta desafíos como la calidad de datos y sesgos, se deben abordar aspectos éticos y técnicos. La privacidad, seguridad y formación médica adecuada son esenciales Conclusión: La IA transforma la salud al mejorar el diagnóstico, personalización de tratamientos y acceso a la atención médica. Los desafíos deben abordarse para aprovechar su potencial en beneficio de los pacientes y la sociedad.
... An algorithm trained on mostly Caucasian patients is not expected to have the same accuracy when applied to minorities [5]. Rigorous evaluation and re-calibration must be done to capture those patient demographics that change over time [6]. Healthcare, with its abundance of data, is, in theory, well-poised to benefit from growth in cloud computing. ...
... The largest and most valuable store of data in healthcare rests in Electronic Medical Records (EMR). However, clinicians' satisfaction with EMRs remains low, resulting in variable completeness and data entry quality, and provider interoperability remains elusive [6]. The typical lament of clinicians is still, "Why is my EMR still inaccurate, and why don't all these systems just talk to each other?". ...
Article
Full-text available
Upon a diagnosis, the clinical team faces two main questions: what treatment, and at what dose? Clinical trials' results provide the basis for guidance and support for official protocols that clinicians use to base their decisions. However, individuals do not consistently demonstrate the reported response from relevant clinical trials. The decision complexity increases with combination treatments where drugs administered together can interact with each other, which is often the case. Additionally, the individual's response to the treatment varies with the changes in their condition. In practice, the drug and the dose selection depend significantly on the medical protocol and the medical team's experience. As such, the results are inherently varied and often suboptimal. Big data and Artificial Intelligence (AI) approaches have emerged as excellent decision-making tools, but multiple challenges limit their application. AI is a rapidly evolving and dynamic field with the potential to revolutionize various aspects of human life. AI has become increasingly crucial in drug discovery and development. AI enhances decision-making across different disciplines, such as medicinal chemistry, molecular and cell biology, pharmacology, pathology, and clinical practice. In addition to these, AI contributes to patient population selection and stratification. The need for AI in healthcare is evident as it aids in enhancing data accuracy and ensuring the quality care necessary for effective patient treatment. AI is pivotal in improving success rates in clinical practice. The increasing significance of AI in drug discovery, development, and clinical trials is underscored by many scientific publications. Despite the numerous advantages of AI, such as enhancing and advancing Precision Medicine (PM) and remote patient monitoring, unlocking its full potential in healthcare requires addressing fundamental concerns. These concerns include data quality, the lack of well-annotated large datasets, data privacy and safety issues, biases in AI algorithms, legal and ethical challenges, and obstacles related to cost and implementation. Nevertheless, integrating AI in clinical medicine will improve diagnostic accuracy and treatment outcomes, contribute to more efficient healthcare delivery, reduce costs, and facilitate better patient experiences, making healthcare more sustainable. This article reviews AI applications in drug development and clinical practice, making healthcare more sustainable, and highlights concerns and limitations in applying AI.
... 6 Medical Device Software (MDSW) encompasses a broad range of applications that support clinical decisions, offering recommendations for diagnosis, prognosis, monitoring, and treatment. These include advanced tools for analyzing radiology images, 12 oncology software supporting genetic analysis, 13 ophthalmology solutions for image recognition, 14 and systems that assist in general medical decisionmaking. 15 Increasingly, machine learning (ML) models are deployed in areas like diabetes management 16 and tuberculosis diagnosis, 17 with studies showing that computer-aided MDSW can sometimes surpass human accuracy in certain diagnostic tasks, such as detecting tuberculosis and diabetic retinopathy. ...
Preprint
Full-text available
Artificial intelligence (AI) in medical device software (MDSW) represents a transformative clinical technology, attracting increasing attention within both the medical community and the regulators. In this study, we leverage a data-driven approach to automatically extract and analyze AI-enabled medical devices (AIMD) from the National Medical Products Administration (NMPA) regulatory database. The continued increase in publicly available regulatory data requires scalable methods for analysis. Automation of regulatory information screening is essential to create reproducible insights that can be quickly updated in an ever changing medical device landscape. More than 4 million entries were assessed, identifying 2,174 MDSW registrations, including 531 standalone applications and 1,643 integrated within medical devices, of which 43 were AI-enabled. It was shown that the leading medical specialties utilizing AIMD include respiratory (20.5%), ophthalmology/endocrinology (12.8%), and orthopedics (10.3%). This approach greatly improves the speed of data extracting providing a greater ability to compare and contrast. This study provides the first extensive, data-driven exploration of AIMD in China, showcasing the potential of automated regulatory data analysis in understanding and advancing the landscape of AI in medical technology.
... For example, radiology software is used to screen and diagnose large amounts of X-ray images [1]. A combined AI and ML approach can also be applied in, for example, oncology for the next-generation sequencing [2], in ophthalmology for image recognition [3], or as a support system for general medical decision-making [4]. ML models have been used for anything from improving outcomes for diabetic patients [5] to tuberculosis diagnosis [6]. ...
Article
Full-text available
The China State Council released the new generation artificial intelligence (AI) development plan, outlining China's ambitious aspiration to assume global leadership in AI by the year 2030. This initiative underscores the extensive applicability of AI across diverse domains, including manufacturing, law, and medicine. With China establishing itself as a major producer and consumer of medical devices, there has been a notable increase in software registrations. This study aims to study the proliferation of health care–related software development within China. This work presents an overview of the Chinese regulatory framework for medical device software. The analysis covers both software as a medical device and software in a medical device. A comparative approach is employed to examine the regulations governing medical devices with AI and machine learning in China, the United States, and Europe. The study highlights the significant proliferation of health care–related software development within China, which has led to an increased demand for comprehensive regulatory guidance, particularly for international manufacturers. The comparative analysis reveals distinct regulatory frameworks and requirements across the three regions. This paper provides a useful outline of the current state of regulations for medical software in China and identifies the regulatory challenges posed by the rapid advancements in AI and machine learning technologies. Understanding these challenges is crucial for international manufacturers and stakeholders aiming to navigate the complex regulatory landscape.
... Additionally, decision aids assist patients in making informed healthcare decisions by offering detailed information about treatment options and outcomes (110,111). AI is utilized for evidence-based clinical decision support (112), detecting adverse events, and using electronic health record (EHR) data to predict patients at risk of readmission (113). By accessing EHR data, AI has demonstrated potential to surpass physicians in diagnostic accuracy (114)(115)(116)(117). Algorithms that combine imaging and EHR data with relevant medical records can predict malignancy on biopsy and differentiate between normal and abnormal screening results, significantly reducing missed breast cancer diagnoses (118). ...
Article
Full-text available
Precision medicine, characterized by the personalized integration of a patient’s genetic blueprint and clinical history, represents a dynamic paradigm in healthcare evolution. The emerging field of personalized anesthesia is at the intersection of genetics and anesthesiology, where anesthetic care will be tailored to an individual’s genetic make-up, comorbidities and patient-specific factors. Genomics and biomarkers can provide more accurate anesthetic protocols, while artificial intelligence can simplify anesthetic procedures and reduce anesthetic risks, and real-time monitoring tools can improve perioperative safety and efficacy. The aim of this paper is to present and summarize the applications of these related fields in anesthesiology by reviewing them, exploring the potential of advanced technologies in the implementation and development of personalized anesthesia, realizing the future integration of new technologies into clinical practice, and promoting multidisciplinary collaboration between anesthesiology and disciplines such as genomics and artificial intelligence.
Article
AI is notably overcoming the long-standing problems, such as high costs, prolonged timelines, and complex biological data analysis, in drug discovery, which in turn is revolutionising the pharmaceutical industry. This article is aimed at making readers realise the significance of AI in drug discovery and what actual changes it has triggered by innovating in areas of target identification, virtual screening, automated drug design, compound optimisation, and biomarker discovery. AI applications, like deep learning or generative models, are now moving much faster and are more accurate in the identification of potential drug targets, while AI-powered virtual screening is the advanced method that makes possible lead identification by the prediction of ligand-receptor binding affinities. For automated drug design tools, generative adversarial networks (GANs) are used for optimising the properties of new molecules, thereby producing the most effective drugs, and reinforcement learning allows the reduction of the possible side effects to further improve the quality of compounds. Biomarker discovery, which is powered by AI, helps in precision medicine by allowing patient stratification and optimisation of clinical trials. Nevertheless, the difficulties in this matter are still poignant. Data handling, transparency in models, regulation uncertainties, and ethical problems such as privacy and bias limit AI in drug development. Collaboration of data sharing among the organisations and the progress in the regulatory frameworks are the most important points to be addressed to solve these issues. Despite these drawbacks, the future of artificial intelligence applications is quite bright, showing possibilities to decrease the spending on R&D, cut the timelines for drug development, deliver precision medicines that improve patients' outcomes, and spur the world's global healthcare solutions.
Article
Full-text available
Objective To analyze a glioblastoma tumor specimen with 3 different platforms and compare potentially actionable calls from each. Methods Tumor DNA was analyzed by a commercial targeted panel. In addition, tumor-normal DNA was analyzed by whole-genome sequencing (WGS) and tumor RNA was analyzed by RNA sequencing (RNA-seq). The WGS and RNA-seq data were analyzed by a team of bioinformaticians and cancer oncologists, and separately by IBM Watson Genomic Analytics (WGA), an automated system for prioritizing somatic variants and identifying drugs. Results More variants were identified by WGS/RNA analysis than by targeted panels. WGA completed a comparable analysis in a fraction of the time required by the human analysts. Conclusions The development of an effective human-machine interface in the analysis of deep cancer genomic datasets may provide potentially clinically actionable calls for individual patients in a more timely and efficient manner than currently possible. ClinicalTrials.gov identifier NCT02725684.
Article
Full-text available
Life sciences researchers are under pressure to innovate faster than ever. Big data offer the promise of unlocking novel insights and accelerating breakthroughs. Ironically, although more data are available than ever, only a fraction is being integrated, understood, and analyzed. The challenge lies in harnessing volumes of data, integrating the data from hundreds of sources, and understanding their various formats. New technologies such as cognitive computing offer promise for addressing this challenge because cognitive solutions are specifically designed to integrate and analyze big datasets. Cognitive solutions can understand different types of data such as lab values in a structured database or the text of a scientific publication. Cognitive solutions are trained to understand technical, industry-specific content and use advanced reasoning, predictive modeling, and machine learning techniques to advance research faster. Watson, a cognitive computing technology, has been configured to support life sciences research. This version of Watson includes medical literature, patents, genomics, and chemical and pharmacological data that researchers would typically use in their work. Watson has also been developed with specific comprehension of scientific terminology so it can make novel connections in millions of pages of text. Watson has been applied to a few pilot studies in the areas of drug target identification and drug repurposing. The pilot results suggest that Watson can accelerate identification of novel drug candidates and novel drug targets by harnessing the potential of big data.
Article
Full-text available
Background: Molecularly targeted agents have been reported to have anti-tumour activity for patients whose tumours harbour the matching molecular alteration. These results have led to increased off-label use of molecularly targeted agents on the basis of identified molecular alterations. We assessed the efficacy of several molecularly targeted agents marketed in France, which were chosen on the basis of tumour molecular profiling but used outside their indications, in patients with advanced cancer for whom standard-of-care therapy had failed. Methods: The open-label, randomised, controlled phase 2 SHIVA trial was done at eight French academic centres. We included adult patients with any kind of metastatic solid tumour refractory to standard of care, provided they had an Eastern Cooperative Oncology Group performance status of 0 or 1, disease that was accessible for a biopsy or resection of a metastatic site, and at least one measurable lesion. The molecular profile of each patient's tumour was established with a mandatory biopsy of a metastatic tumour and large-scale genomic testing. We only included patients for whom a molecular alteration was identified within one of three molecular pathways (hormone receptor, PI3K/AKT/mTOR, RAF/MEK), which could be matched to one of ten regimens including 11 available molecularly targeted agents (erlotinib, lapatinib plus trastuzumab, sorafenib, imatinib, dasatinib, vemurafenib, everolimus, abiraterone, letrozole, tamoxifen). We randomly assigned these patients (1:1) to receive a matched molecularly targeted agent (experimental group) or treatment at physician's choice (control group) by central block randomisation (blocks of size six). Randomisation was done centrally with a web-based response system and was stratified according to the Royal Marsden Hospital prognostic score (0 or 1 vs 2 or 3) and the altered molecular pathway. Clinicians and patients were not masked to treatment allocation. Treatments in both groups were given in accordance with the approved product information and standard practice protocols at each institution and were continued until evidence of disease progression. The primary endpoint was progression-free survival in the intention-to-treat population, which was not assessed by independent central review. We assessed safety in any patients who received at least one dose of their assigned treatment. This trial is registered with ClinicalTrials.gov, number NCT01771458. Findings: Between Oct 4, 2012, and July 11, 2014, we screened 741 patients with any tumour type. 293 (40%) patients had at least one molecular alteration matching one of the 10 available regimens. At the time of data cutoff, Jan 20, 2015, 195 (26%) patients had been randomly assigned, with 99 in the experimental group and 96 in the control group. All patients in the experimental group started treatment, as did 92 in the control group. Two patients in the control group received a molecularly targeted agent: both were included in their assigned group for efficacy analyses, the patient who received an agent that was allowed in the experimental group was included in the experimental group for the purposes of safety analyses, while the other patient, who received a molecularly targeted agent and chemotherapy, was kept in the control group for safety analyses. Median follow-up was 11·3 months (IQR 5·8-11·6) in the experimental group and 11·3 months (8·1-11·6) in the control group at the time of the primary analysis of progression-free survival. Median progression-free survival was 2·3 months (95% CI 1·7-3·8) in the experimental group versus 2·0 months (1·8-2·1) in the control group (hazard ratio 0·88, 95% CI 0·65-1·19, p=0·41). In the safety population, 43 (43%) of 100 patients treated with a molecularly targeted agent and 32 (35%) of 91 patients treated with cytotoxic chemotherapy had grade 3-4 adverse events (p=0·30). Interpretation: The use of molecularly targeted agents outside their indications does not improve progression-free survival compared with treatment at physician's choice in heavily pretreated patients with cancer. Off-label use of molecularly targeted agents should be discouraged, but enrolment in clinical trials should be encouraged to assess predictive biomarkers of efficacy.
Article
Full-text available
The recent FDA approval of the MiSeqDx platform provides a unique opportunity to develop targeted next generation sequencing (NGS) panels for human disease, including cancer. We have developed a scalable, targeted panel-based assay termed UNCseq, which involves a NGS panel of over 200 cancer-associated genes and a standardized downstream bioinformatics pipeline for detection of single nucleotide variations (SNV) as well as small insertions and deletions (indel). In addition, we developed a novel algorithm, NGScopy, designed for samples with sparse sequencing coverage to detect large-scale copy number variations (CNV), similar to human SNP Array 6.0 as well as small-scale intragenic CNV. Overall, we applied this assay to 100 snap-frozen lung cancer specimens lacking same-patient germline DNA (07-0120 tissue cohort) and validated our results against Sanger sequencing, SNP Array, and our recently published integrated DNA-seq/RNA-seq assay, UNCqeR, where RNA-seq of same-patient tumor specimens confirmed SNV detected by DNA-seq, if RNA-seq coverage depth was adequate. In addition, we applied the UNCseq assay on an independent lung cancer tumor tissue collection with available same-patient germline DNA (11-1115 tissue cohort) and confirmed mutations using assays performed in a CLIA-certified laboratory. We conclude that UNCseq can identify SNV, indel, and CNV in tumor specimens lacking germline DNA in a cost-efficient fashion.
Article
Full-text available
Identifying somatic mutations is critical for cancer genome characterization and for prioritizing patient treatment. DNA whole exome sequencing (DNA-WES) is currently the most popular technology; however, this yields low sensitivity in low purity tumors. RNA sequencing (RNA-seq) covers the expressed exome with depth proportional to expression. We hypothesized that integrating DNA-WES and RNA-seq would enable superior mutation detection versus DNA-WES alone. We developed a first-of-its-kind method, called UNCeqR, that detects somatic mutations by integrating patient-matched RNA-seq and DNA-WES. In simulation, the integrated DNA and RNA model outperformed the DNA-WES only model. Validation by patient-matched whole genome sequencing demonstrated superior performance of the integrated model over DNA-WES only models, including a published method and published mutation profiles. Genome-wide mutational analysis of breast and lung cancer cohorts (n = 871) revealed remarkable tumor genomics properties. Low purity tumors experienced the largest gains in mutation detection by integrating RNA-seq and DNA-WES. RNA provided greater mutation signal than DNA in expressed mutations. Compared to earlier studies on this cohort, UNCeqR increased mutation rates of driver and therapeutically targeted genes (e.g. PIK3CA, ERBB2 and FGFR2). In summary, integrating RNA-seq with DNA-WES increases mutation detection performance, especially for low purity tumors.
Article
This Viewpoint argues against universal tumor sequencing.Is there enough benefit to justify sequencing all patients’ tumors? No.Article InformationCorresponding Author: Howard (Jack) West, MD, Swedish Cancer Institute, 1221 Madison St, Ste 200, Seattle, WA 98104 (howard.west@swedish.org).Published Online: April 14, 2016. doi:10.1001/jamaoncol.2016.0075.Conflict of Interest Disclosures: None reported.
Article
This Viewpoint argues for universal tumor sequencing.Is there enough benefit to justify sequencing all patients’ tumors? Yes.Article InformationCorresponding Author: Razelle Kurzrock, MD, Moores Cancer Center, University of California–San Diego, 3855 Health Sciences Dr, MC 0658, La Jolla, CA 92093-0658 (rkurzrock@ucsd.edu).Published Online: April 14, 2016. doi:10.1001/jamaoncol.2016.0078.Conflict of Interest Disclosures: Dr Subbiah reports research funds from Novartis, Bayer, GSK, Nanocarrier, Vegenics, Northwest Biotherapeutics, Berghealth, Incyte, Fujifilm, Pharmamar, D3, Pfizer, Amgen, and Abbvie. Dr Kurzrock has research funding from Genentech, Merck Serono, Pfizer, Sequenom, Foundation Medicine, and Guardant; consultant fees from Sequenom and Actuate Therapeutics; and an ownership interest in Novena Inc and Curematch Inc. No other disclosures are reported.
Conference Paper
Purpose: Assistance algorithms for medical tasks have great potential to support physicians with their daily work. However, medicine is also one of the most demanding domains for computer-based support systems, since medical assistance tasks are complex and the practical experience of the physician is crucial. Recent developments in the area of cognitive computing appear to be well suited to tackle medicine as an application domain. Methods: We propose a system based on the idea of cognitive computing and consisting of auto-configurable medical assistance algorithms and their self-adapting combination. The system enables automatic execution of new algorithms, given they are made available as Medical Cognitive Apps and are registered in a central semantic repository. Learning components can be added to the system to optimize the results in the cases when numerous Medical Cognitive Apps are available for the same task. Our prototypical implementation is applied to the areas of surgical phase recognition based on sensor data and image progressing for tumor progression mappings. Results: Our results suggest that such assistance algorithms can be automatically configured in execution pipelines, candidate results can be automatically scored and combined, and the system can learn from experience. Furthermore, our evaluation shows that the Medical Cognitive Apps are providing the correct results as they did for local execution and run in a reasonable amount of time. Conclusion: The proposed solution is applicable to a variety of medical use cases and effectively supports the automated and self-adaptive configuration of cognitive pipelines based on medical interpretation algorithms.
Article
The necessary infrastructure to carry out genomics-driven oncology is now widely available and has resulted in the exponential increase in characterized cancer genomes. While a subset of genomic alterations is clinically actionable, the majority of somatic events remain classified as variants of unknown significance and will require functional characterization. A careful cataloging of the genomic alterations and their response to therapeutic intervention should allow the compilation of an "actionability atlas" and the creation of a genomic taxonomy stratified by tumor type and oncogenic pathway activation. The next phase of genomic medicine will therefore require talented bioinformaticians, genomic navigators, and multidisciplinary approaches to decode complex cancer genomes and guide potential therapy. Equally important will be the ethical and interpretable return of results to practicing oncologists. Finally, the integration of genomics into clinical trials is likely to speed the development of predictive biomarkers of response to targeted therapy as well as define pathways to acquired resistance.