ArticlePDF AvailableLiterature Review

Genomic Medicine: A Decade of Successes, Challenges, and Opportunities

Authors:
  • Precision Medicine Advisors

Abstract and Figures

Genomic medicine-an aspirational term 10 years ago-is gaining momentum across the entire clinical continuum from risk assessment in healthy individuals to genome-guided treatment in patients with complex diseases. We review the latest achievements in genome research and their impact on medicine, primarily in the past decade. In most cases, genomic medicine tools remain in the realm of research, but some tools are crossing over into clinical application, where they have the potential to markedly alter the clinical care of patients. In this State of the Art Review, we highlight notable examples including the use of next-generation sequencing in cancer pharmacogenomics, in the diagnosis of rare disorders, and in the tracking of infectious disease outbreaks. We also discuss progress in dissecting the molecular basis of common diseases, the role of the host microbiome, the identification of drug response biomarkers, and the repurposing of drugs. The significant challenges of implementing genomic medicine are examined, along with the innovative solutions being sought. These challenges include the difficulty in establishing clinical validity and utility of tests, how to increase awareness and promote their uptake by clinicians, a changing regulatory and coverage landscape, the need for education, and addressing the ethical aspects of genomics for patients and society. Finally, we consider the future of genomics in medicine and offer a glimpse of the forces shaping genomic medicine, such as fundamental shifts in how we define disease, how medicine is delivered to patients, and how consumers are managing their own health and affecting change.
Content may be subject to copyright.
GENOMICS
Genomic Medicine: A Decade of Successes, Challenges,
and Opportunities
Jeanette J. McCarthy,
1,2
Howard L. McLeod,
3
Geoffrey S. Ginsburg
1
*
Genomic medicinean aspirational term 10 years agois gaining momentum across the entire clinical continuum
from risk assessment in healthy individuals to genome-guided treatment in patients with complex diseases. We
review the latest achievements in genome research and their impact on medicine, primarily in the past decade.
In most cases, genomic medicine tools remain in the realm of research, but some tools are crossing over into clinical
application, where they have the potential to markedly alter the clinical care of patients. In this State of the Art
Review, we highlight notable examples including the use of next-generation sequencing in cancer pharmaco-
genomics, in the diagnosis of rare disorders, and in the tracking of infectious disease outbreaks. We also discuss
progress in dissecting the molecular basis of common diseases, the role of the host microbiome, the identification
of drug response biomarkers, and the repurposing of drugs. The significant challenges of implementing genomic
medicine are examined, along with the innovative solutions being sought. These challenges include the difficulty in
establishing clinical validity and utility of tests, how to increase awareness and promote their uptake by clinicians, a
changing regulatory and coverage landscape, the need for education, and addressing the ethical aspects of ge-
nomics for patients and society. Finally, we consider the future of genomics in medicine and offer a glimpse of
the forces shaping genomic medicine, such as fundamental shifts in how we define disease, how medicine is
delivered to patients, and how consumers are managing their own health and affecting change.
INTRODUCTION
This year marks the 10th anniversary of the official completion of the
HumanGenomeProject,aprojectthatenabledthesystematicexplo-
ration of the molecular underpinnings of disease and generated expec-
tations of the transformation of medicine. Although the previous
decades were marked by improvements in overall health and longev-
ity, before completion of the Human Genome Project, the tools used
for diagnosis and treatment of disease were often blunt and imprecise.
Most diseases were defined by anatomical location and clinical symp-
toms and treated with one-size-fits-all therapies that failed to account
for the unique biological background of the individual. Despite the
development of rational drug design strategies, therapeutic efficacy
has remained unacceptably low (1).The Human Genome Project laid
the foundation for genomic medicine, offering a means of defining
disease at the molecular level. Along with advances in genotyping
and sequencing technologies, bioinformatics, systems biology, and
computational biology, the fruits of the Human Genome Project
have fueled important biological discoveries at an unprecedented
rate. Today, genomic medicine aims to build on this foundation,
translating these discoveries into clinical practice, with the ultimate
goal of personalized medicine.
This State of the Art Review highlights the latest achievements in
genome research and their impact on medicine, primarily in the past
decade. In some cases, genomic discoveries have led to marked changes
in the clinical care of patients. However, the transition to genomic med-
icine has not been smooth, and many challenges still exist. We explore
these challenges and discuss innovative solutions. Finally, we contem-
plate the future, offering a glimpse of the expected changes in health
care as genomic medicine takes hold.
GENOMIC MEDICINE SUCCESSES
The term genomic medicinewas virtually absent from our lexicon
before 1995; now, it permeates the medical literature, the press, and
the economy. The Human Genome Project resulted in the launch of
thousands of biotechnology firms globally and the development of
faster and cheaper technologies for querying the genome, which have
fueled discoveries of disease biomarkers and drug targets. The phar-
maceutical industry has adopted genome-enabled drug discovery, and
the market for molecular diagnostics has grown rapidly. There are
now groundbreaking examples of the application of genomics across
the stages of disease, from risk stratification and screening to diagnosis
and prognosis to treatment (Table 1).Forsomediseaseslikebreast
cancer, HIV infection, and chronic hepatitis C virus (HCV) infection,
genome-based tools have been woven into clinical practice and disease
management (Fig. 1), but for most areas of medicine, the uptake of
genomics has been slow.
SEQUENCING TECHNOLOGY: THE DRIVER OF
GENOMIC MEDICINE
Technology has been an enabling force, providing transformative
tools for genomic research including genome-wide association studies
(GWAS) and next-generation sequencing (NGS). In 2005, GWAS
made their debut with the identification of a major susceptibility gene
for a complex trait. Through evaluation of hundreds of common ge-
netic variants, Klein et al. identified mutations in the complement
factor H gene as a genetic cause of age-related macular degeneration
1
Institute for Genome Sciences & Policy, Duke University, Durham, NC 27708, USA.
2
De-
partment of Epidemiology and Biostatistics, UCSF School of Medicine, San Francisco,
CA 94105, USA.
3
Institute for Pharmacogenomics and Individualized Therapy, Univer-
sity of North Carolina, Chapel Hill, NC 27599, USA.
*Corresponding author. E-mail: Geoffrey.ginsburg@duke.edu
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 1
(2). Notwithstanding its limitations, including its restriction to com-
mon variants, incomplete genome coverage, and inherent challenge of
discerning the actual causal genetic variant, GWAS has been a trans-
formative technology, representing a major advance over the one-
gene-at-a-time candidate gene approach used for decades before.
Moreover, GWAS provided an indirect means of querying common
genetic variation across the entire human genome in an unbiased fash-
ion, offering an unprecedented opportunity to uncover new biological
pathways of disease.
From 2001 to 2012, the cost to sequence a human genome
dropped from $100 million to less than $10,000, and these costs con-
tinue to decline (3). Current sequencing machines can read about
250 billion bases in a week compared to only about 5 million in
2000 (4). NGS technology (also known as massively parallel se-
quencing) now allows direct measurement of not just common var-
iants but theoretically all variation in a genome (5). Data from the
1000 Genomes Project have confirmed previous estimates of the
population frequency of germline variants to be about 1 in every
1000 of the 3.2 billion nucleotide positions, giving rise to about
3 million variants in the human genome (6). The effect of genetic
variants in the ~1% of the genome that codes for genes is somewhat
predictable. The challenge lies in figuring out the meaning of var-
iants that occur in the vast remaining noncoding regions of the
genome, the so-called dark matter, whose function is largely unknown.
Consequently, the National Human Genome Research Institute
initiated project ENCODE, the Encyclopedia of DNA Elements,
whose goal is to functionally annotate noncoding regions of the
genome (7). This ongoing initiative will provide new insights into
the organization and regulation of genes throughout the genome,
enhancing the ability to annotate variants of unknown significance.
In the interim, researchers have focused on exome sequencing,
which examines variation in the coding sequence (genes), where
mutations have predictable effects on downstream protein struc-
ture. Individuals typically carry several hundred rare and potentially
deleterious coding region variants (6). For rare, Mendelian disorders,
those controlled by a single gene with a simple pattern of inheritance,
the variants found in as few as one or two affected individuals can
be compared to those found in unaffected individuals to pinpoint
disease-causing mutations. The first successful applications of exome
sequencing came in 2009 with the diagnosis of patients with Freeman-
Sheldon syndrome (8), Miller syndrome (9), and congenital chloride
diarrhea (10). These and other Mendelian disorders, with their rela-
tively simple genetic basis, are amenable to exome sequencing of their
DNA and the DNA of a small number of other affected or unaffected
individuals.
Today, GWAS and NGS are integral tools in basic genomic re-
search, but are increasingly being explored for clinical applications, in-
cluding the clinical diagnosis of rare genetic diseases (11), the selection
of cancer treatments based on molecular characterization of the tumor
(12), and the tracking of infectious disease outbreaks in real time (13).
Elucidating the genetic basis of common diseases has been more
challenging, but research in this area has broadened our understand-
ing of underlying disease mechanisms and has revealed new therapeu-
tic approaches through repurposing of existing drugs for treating
diseases they were not originally intended to treat (1416).
TUMOR SEQUENCING FOR CANCER PHARMACOGENOMICS
One of the most high-profile disease areas to benefit from NGS is
cancer. All cancers arise as a result of DNA mutations, which con-
fer a growth advantage upon the cells in which they have occurred,
giving rise to tumors. Comparison of the genetic profiles of tumors
and the surrounding normal tissue can reveal the acquired changes
driving growth that may be targets for treatment. Meanwhile, com-
parisons across patients and tumor types are changing how cancers
are classified and treated.
Targeted therapies for cancer
The idea of pairing medicines with specific tumor markers in a tar-
geted fashion to improve efficacy of cancer therapies is not new. In the
mid-1980s, detailed molecular studies of breast tumors led to the dis-
covery of HER-2, a biomarker overexpressed in about 30% of breast
tumors and associated with adverse outcomes (17). HER-2 typing of
primary breast tumors provided clinicians with a new tool that could
be used to guide adjuvant chemotherapy (18). The development of
trastuzumab (Herceptin) in 1998, a humanized monoclonal antibody
targeting HER-2, resulted in widespread adoption of the HER-2 test
Table 1. Examples of genomic tests used in clinical practice.
Susceptibility to common diseases
BRCA1/BRCA2 for breast and ovarian cancer predisposition
Lynch syndrome genetic screening in colorectal cancer families
Human leukocyte antigen (HLA) typing to aid in diagnosis of celiac disease
*NGS for Mendelian disorders and diagnostic dilemmas
*NGS for noninvasive prenatal screening/diagnosis
Preclinical diagnosis and prognosis
Oncotype DX: 21-gene RNA signature for breast tumors; 12-gene RNA
signature for colon tumors
MammaPrint: 70-gene RNA signature for breast tumors
OVA1: 5-protein signature for ovarian mass malignancy
AlloMap: 11-gene (blood RNA) signature for monitoring rejection after
cardiac transplant
Corus CAD: 23-gene (blood RNA) signature for coronary artery disease
Cancer pharmacogenomics
HER-2trastuzumab
EGFRgefitinib
KRAScetuximab and panitumumab
ALKcrizotinib
BRAFvemurafenib
Pharmacogenomic dosing
CYP2D9/VKORC1warfarin
Pharmacogenomic adverse events
HLA-B*5701abacavir (HIV infection)
HLA-B*1502carbamazepine (epilepsy, bipolar disorder)
Pharmacogenomic efficacy
CYP2C19clopidogrel (coronary artery disease, peripheral vascular disease)
IL28Bpegylated interferon/ribavirin (HCV infection)
*NGS, next-generation sequencing.
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 2
(19), now part of the standard workup and management of breast
cancer (2022) (Fig. 2). During the following decade, other examples
of cancer therapies with companion diagnostics emerged (Table 1).
The introduction of EGFR mutation testing has markedly improved
the efficacy of gefitinib and erlotinib, small-molecule drugs for the
treatment of nonsmall cell lung cancer that target epidermal growth
factor receptor (EGFR) (23,24). In metastatic colorectal cancer, tu-
mors with mutated KRAS are almost always resistant to treatment with
cetuximab and panitumumab, leading the American Society of Clinical
Oncologists and the U.S. Food and Drug Administration (FDA) to
recommend withholding the drugs in these patients (25,26). With
the introduction of NGS, researchers and clinicians can now make a
comprehensive assessment of tumor markers, increasing the rate of
discovery of new targets, new therapeutic alternatives, and new clinical
management guidelines for cancer treatment. In 2011, two cancer
drugs received accelerated approval by the FDA for use with a com-
panion diagnostic test: crizotinib for the treatment of patients with lo-
cally advanced or metastatic nonsmall cell lung cancer with its
companion diagnostic designed to detect the EML4-ALK fusion gene
(27) and vemurafenib for the treatment of patients with metastatic or
unresectable melanoma positive for BRAF V600E mutations (28).
More recently, NGS of tumors has identified the existence of the
same mutations in distinct cancer types, prompting the expansion of
indications for some FDA-approved drugs. BRAF V600E mutations
not only are common in melanoma but also have been widely ob-
served in other cancers (29), especially hairy cell leukemia (30), leading
to the expanded use and ultimate demonstration of vemurafenib as a
viable treatment option for refractory hairy cell leukemia (31). Simi-
larly, early studies indicate that crizotinib, targeting EML4-ALKpositive
nonsmall cell lung cancers, is effective against other types of tumors
containing ALK alterations, such as aggressive forms of pediatric neu-
roblastoma and anaplastic large cell lymphoma (32). As the field pro-
gresses, the way tumors are classified is shifting away from tissue of
origin and toward molecular taxonomy, having a profound impact
on the manner in which treatment decisions are made.
Tumor genome analysis is also being explored in cancer patients to
identify new markers and mechanisms of drug sensitivity or resistance.
By sequencing the tumor of a patient with durable remission of meta-
static bladder cancer after treatment with everolimus, Iyer et al. identi-
fied a loss-of-function mutation in TSC1 as a marker of everolimus
sensitivity (33). Similarly, patients with nonsmall cell lung cancer
treated with crizotinib are prone to relapse, leading to the development
of next-generation ALK inhibitors to overcome crizotinib resistance.
Genomic alterations in resistant tumors were found to correlate with
response to these newer therapies, providing the rationale for pursuing
targeted combinatorial therapeutics to combat resistance (34).
NGS of tumors is not without its limitations (35). Tissue sampling
is problematic: The limited availability of tissue from a standard
biopsy, the need for fresh (as opposed to preserved) tissue, the inher-
ent heterogeneity within a tumor, the presence of aneuploidy, and the
contamination of tumor samples with surrounding normal tissue all
affect the ability to comprehensively characterize tumor mutations.
Further computational and experimental approaches are required to
distinguish those mutations responsible for cancerous growth (drivers)
from those that are inconsequential (passengers). The International
Cancer Genome Consortium (https://www.icgc.org/icgc) and the Cancer
Genome Atlas (http://cancergenome.nih.gov/) represent international
collaborative efforts to define the spectrum of mutations found in tumors,
mapping the genomic landscape of cancer. These efforts will provide a
foundation from which to develop therapeutic strategies against new tar-
gets, but this process is not trivial and, even when successful, may be short-
lived as therapeutic resistance evolves. Thus, whereas NGS is a promising
new tool, it is not a panacea for cancer genomic medicine.
Circulating tumor markers
Another noteworthy application of NGS is noninvasive cancer detec-
tion, either in the setting of primary diagnosis or in monitoring recur-
rence after treatment. Leary and colleagues have laid the groundwork
for this, demonstrating that chromosomal rearrangements, a hallmark
of cancerous tumors, can be detected in blood samples from cancer
patients using NGS (36). This method is successful at discriminating
samplesfromcancerpatientsfromthose of healthy controls, but the
sensitivity and specificity depend highly on the amount of tumor DNA
present in the sample and the detection limits of the sequencing technol-
ogy (37). This method shows promise for monitoring tumor burden to
determine response to treatment, providing an alternative to serial radio-
graphic imaging, which often fails to detect changes in tumor burden. In
metastatic breast cancer, Dawson and colleagues demonstrated marked
improvements in detection of chromosomal rearrangements as markers
of tumor burden, over and beyond specific cancer antigens or generic
circulating tumor DNA (38). Improvements in NGS technology will only
enhance the ability to detect and monitor cancer, with the potential to
markedly alter how the disease is managed.
HCV
HIV
Breast cancer
Disease burdenDisease burden Disease burden
BRCA1/2 Oncotype DX
MammaPrint HER-2: Herceptin
Viral RNA load HLA: Abacavir
Viral RNA load
Viral genotype
IL28B: PegIFN
efficacy and dosing
ITPA: Ribavirin
adverse events
BRCA
BRCA
BRCA
BRCA
BRCA
BRCA
BRCA
1/2
1/2
1/2
1/2
2
BRCA1/2
Onco
Onco
Onco
nc
type
type
type
type
type
type
e
e
DX DX
DX
DX
DX
DX
X
Mamm
Mamm
Mamm
Mamm
Mamm
Mamm
Ma
aPri
aPri
aPri
aPri
aPri
aPr
a
t
nt
nt
nt
nt
nt
t
Oncotype DX
MammaPrint
HER
HER-
HER-
HER-
HER-
HER-
R
2: H
2: H
2: H
2: H
2: H
H
erce
erce
erce
erce
erce
erce
erce
ptin
ptin
ptin
ptin
ptin
ptin
t
HER-2: Herceptin
Vira
Vira
Vira
Vira
Vira
Vira
a
lRN
l RN
l RN
l RN
l RN
l RN
l RN
Alo
A lo
A lo
A lo
A lo
A lo
A
ad
ad
ad
ad
ad
ad
ad
Viral RNA load
HLA:
HLA:
HLA:
HLA:
HLA:
HLA
A
Aba
Aba
Aba
Aba
Aba
Aba
Aba
cavi
cavi
cavi
cavi
cavi
cav
cav
r
r
r
r
r
r
HLA: Abacavir
Vira
Vira
Vira
Vira
Vira
Vira
a
lRN
l RN
l RN
l RN
l RN
l RN
l RN
Alo
A lo
A lo
A lo
A lo
A lo
A
ad
ad
ad
ad
ad
ad
ad
Vira
Vira
Vira
Vira
Vira
Vira
a
l ge
l ge
l ge
l ge
l ge
l ge
l ge
noty
noty
noty
noty
noty
noty
not
pe
pe
pe
pe
pe
pe
e
Viral RNA load
Viral genotype
IL28
IL28
IL28
L28
L28
8
8
B: P
B: P
B: P
B: P
B: P
B: P
B
egIFegIF
egIF
egIF
egIF
egIF
eg
N
N
N
N
N
N
N
effi
effi
effi
effi
effi
effi
effi
cacy
cacycacycacy
cacy
cacy
cac
and
and
and
and
and
and
and
dos
dos
dos
dos
dos
dos
ing
ing
ing
ing
ing
ing
ITPA
ITPA
ITPA
ITPA
TPA
TPA
A
A
:Ri
: Ri
: Ri
: Ri
: Ri
: Ri
R
bavi
bavi
bavi
bavi
bavi
bav
av
rin
rin
rin
rin
rin
n
adve
adve
adve
adve
adve
adve
adve
rse
rse
rse
rse
rse
rse
rse
even
even
even
even
even
even
eve
ts
ts
ts
ts
ts
ts
t
IL28B: PegIFN
efficacy and dosing
ITPA: Ribavirin
adverse events
Health
Time
Preclinical Clinical
Susceptibility Screening Diagnosis Prognosis Treatment
Fig. 1. Genomic medicine in action. Examples of three conditions where
genomic medicine tools and information have affected clinical management
at various stages of disease. (Top) In chronic HCV infection, several genomic
markers are used in disease management: viral RNA concentrations are used
for diagnosis and monitoring; the genotypeofthevirusisusedforprognosis
and treatment response; and host IL28B and ITPA genotypes are used as
pharmacogenomic markers for efficacy and dosing. (Middle) In patients in-
fected with HIV, viral RNA concentrations are used for diagnosis and monitoring,
and host HLA genotype is used as a pharmacogenomic marker of hypersen-
sitivity to abacavir treatment. (Bottom) In breast cancer, germline BRCA1/BRCA2
genotyping is used to determine susceptibility to breast and ovarian cancer;
analysis of tumors with Oncotype Dx or MammaPrint is used to predict like-
lihood of recurrence; and expression of the tumor marker HER-2 is used to
inform treatment with the monoclonal antibody Herceptin.
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 3
CLINICAL SEQUENCING AND DIAGNOSTIC DILEMMAS
According to the Online Mendelian Inheritance in Man database
(http://omim.org/), about 3676 Mendelian disorders have a known
molecular basis. However, there are nearlyasmanysuspectedMendelian
traits for which the molecular basis remains to be identified. The
potential for clinical sequencing to find the underlying cause and iden-
tify treatment options for these rare, debilitating diseases has led to the
formation of various large national and international consortia. In the
United States, three Centers for Mendelian Genomics have been es-
tablished at the University of Washington, Yale University, and a joint
Center at Baylor College of Medicine and Johns Hopkins University
(39). Moreover, clinical sequencing is being offered to patients with
unknown yet suspected genetic diseases, the so-called diagnostic
Diagnosis: molecular taxonomy Treatment: tailored choices
Expanded definition of self
Host germline genome
Tumor genome Microbiome
Infectious agents
Targeting specific disease markers
Cancer cell
HER-2 protein
Herceptin
Improved likelihood of response
HCV patients: PegIFN treatment responders
IL28B genotype CT/TT 40%
80%IL28B genotype CC
Enhanced drug safety
Abacavir hypersensitive
Abacavir tolerant
HLA type
Patients (%)
0
80
HLA-B*5701 HLA-DR7 +
HLA-DQ3
HLA-B*5701 +
HLA-DR7 +
HLA-DQ3
Dosing optimization
iWarfarin app for iPhone
Earlier diagnosis
Clinical
Mendelian
sequencing
Expanded
newborn
screening
Noninvasive
prenatal
testing
Earlier diagnosis in human disease
Disease burden
BRCA1/2 testing
for breast
cancer risk
Health
Corus CAD
test for
heart disease
Time
Preclinical
Oncotype DX
test for ER+
breast cancer
diagnosis
Clinical
Fig. 2. Trends in genomic medicine. Several themes are emerging in
the diagnosis and treatment of disease. (Left) First, as the knowledge of
the genomic underpinnings of disease increases, the definition of self
is likewise expanding. This self includes not just the germline human
genome inherited from our parents but also somatic changes in the ge-
nomes of tumors, the genomes of commensal microbes that inhabit our
body, and the genomes of pathogenic organisms. Second, the influence
of genomics on medicine is moving the diagnosis of diseases toward
the earliest possible point in the course of disease, before clinical man-
ifestation. Third, genomic testing is occurring earlier in the life of a human,
moving from adulthood to childhood, the neonatal period, and even pre-
natally. (Right) Pharmacogenomic markers are being used in several ways:
to select targeted therapies exemplified by the breast cancer marker HER-2
and treatment with the monoclonal antibody Herceptin; to predict likeli-
hood of response, as in the case of the host IL28B genotype and response
to interferon therapy in HCV infection; to enhance drug safety, for example,
by testing for host HLA genotypes predictive of abacavir hypersensitivity in
humans infected with HIV; and to optimize dosing of drugs, such as war-
farin, based on genotypes indicative of rate of metabolism using, for exam-
ple, the iWarfarin app.
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 4
dilemmas, at many major medical centers and through programs
such as the National Human Genome Research InstitutesUndiagnosed
Diseases Program (http://www.genome.gov/27544402). One notable
case from the Medical College of Wisconsin involved the use of clinical
exome sequencing to identify a gene variant in a child who presented
to the clinic with a severe, intractable form of inflammatory bowel dis-
ease for which physicians were unable to provide a diagnostic explana-
tion (40). The discovered variant in the XIAP gene defined a new form
of inflammatory bowel disease due to deficiency of an X-linked inhibitor
of apoptosis. Knowledge of the causal mutation led to a cure through
allogeneic bone marrow transplantation and demonstrated the power
of clinical NGS as a strategy for diagnosis and management of rare
diseases. Early results from a number of clinical sequencing programs
estimate the success rate of disease gene identification at about 50%,
offering hope to thousands of individuals with previously undiagnosed
or untreated rare disorders (41,42).
Newborn screening, prenatal diagnosis, and preconception
carrier testing
A natural outcome of identifying genes for rare Mendelian disorders
is the application of these findings to earlier detection, either at birth
(newborn screening), in utero (prenatal diagnosis), or preconception
(carrier testing). Newborn screening is a mandatory, state-supported
public health program meant to protect newborn children by screen-
ing them for rare, treatable (and thus preventable) disorders at birth.
The scope of diseases tested varies by state but has been steadily
increasing from an average of 5 conditions in 1995 to a uniform pan-
el of 31 core disorders and 26 secondary disorders currently recom-
mended by the U.S. Department of Health and Human Services
Secretarys Advisory Committee on Heritable Disorders in New-
borns and Children (43). Some states evaluate additional conditions,
including diseases that affect children at a later stage in development
and others for which the benefits of early intervention are limited.
The number of conditions considered for newborn screening will
undoubtedly grow with genome sequencing, improving early diag-
nosis. A concern is that, as a consequence, expanded screening will
erode the ethical justification for this compulsory program and may
undermine one of the most successful public health programs in ex-
istence (44).
Prenatal diagnosis moves screening for genetic disorders even fur-
ther upstream by detecting genetic variation in the developing fetus.
Traditionally, fetal DNA is obtained invasively through amniocentesis
or chorionic villus sampling, but a potential alternative emerged in
1997 when it was demonstrated that cell-free fetal DNA circulates in
maternal blood and could be isolated, amplified, and sequenced non-
invasively through a sample of maternal plasma (45). In 2008, NGS
technologies were used successfully for the first time to identify fetal
aneuploidy from cell-free fetal DNA in maternal plasma (46,47). Clin-
ical trials of the new method rapidly followed, and by late 2011,
noninvasive prenatal testing of trisomy 21 by sequencing of maternal
plasma DNA was being offered on a clinical and commercial basis in
the United States and China (48). In the third quarter of 2012, Sequenom
Inc., one such company offering this test, had reached a 90,000 an-
nualized test volume run rate for the MaterniT21 PLUS test (49).
Noninvasive prenatal testing for disorders caused by large dele-
tions, single-nucleotide substitutions, and other types of genetic vari-
ation, as opposed to de novo chromosomal aneuploidies like trisomy
21, is more challenging due to the factthatmaternalplasmacontains
DNA from both the fetus and mother, who may share the same mu-
tation. However, progress has been made in this area as well (50,51).
Investigators at Stanford University applied noninvasive prenatal test-
ing to detect a large genetic deletion underlying DiGeorge syndrome
in a first-trimester fetus using only a blood sample from the mother
(52,53). Noninvasive prenatal testing eliminates the need for invasive
procedures while also greatly expanding the number of genetic var-
iants that have traditionally been detected in utero.
Even before conception, carrier screening enables couples who are
planning a family to assess their risk of having a child with a recessive
Mendelian disorder and use this information to guide their reproduc-
tive decisions. There are more than 1000 rare, recessive Mendelian dis-
orders for which the underlying genetic mutation is known (54). Although
individually rare, Mendelian disorders can have a sizable public health
impact. Population-based carrier screening for Tay-Sachs disease has
reduced the incidence of the disease in Jewish populations in the
United States and Canada by more than 90% (55). Considering that
each person is estimated to carry on average 2.8 mutations for known
severe recessive disorders (56), the impact of screening could be sizable
in terms of reduced disease morbidity and mortality in the population.
The impact could be particularly significant in populations such as
Ashkenazi Jewish, who are disproportionately affected by recessive
disorders. There may also be utility for screening in the setting of in vitro
fertilization, where preimplantation genetic analysis has the potential to
reduce the incidence of disease. Panels of tests for hundreds of child-
hood recessive illnesses with severe clinical manifestations have been
developed (56), and some are offered directly to patients through
companiessuchasCounsylInc.
Genetics of common complex diseases
Unlike rare Mendelian disorders, the genetic dissection of common,
complex diseases such as cancer, cardiovascular disease, and diabetes
has proven to be more difficult. These diseases by definition are due to
the complex interplay of many gene variants, both common and rare,
as well as nongenetic factors. GWAS, the current method used to find
these genes, has been carried out on large cohorts of patients and
controls across numerous traits and diseases, revealing hundreds of
common genetic variants associated with those traits (http://www.
genome.gov/gwastudies). Researchers are now turning to NGS to
identify rare genetic variants associated with complex diseases. For
example, exome sequencing in neurodevelopmental conditions has
identified de novo mutations linked to diseases like autism and schiz-
ophrenia (57). However, exome sequencing of large numbers of subjects
with a complex trait can be costly and experimentally challenging (58).
Often, thousands of patients are required to assign statistical significance
to rare variant associations with complex diseases. Innovative study
designs such as exome sequencing of patients with extreme phenotypes
have yielded some successes, including the identification of DCTN4
associated with clinical sequelae in cystic fibrosis patients infected with
Pseudomonas aeruginosa (59).
Despite the unequivocally strong statistical associations between
genetic variants and complex diseases, their low sensitivity and spec-
ificity afford limited clinical value for disease predisposition testing.
Notable exceptions include genes underlying breast cancer (60), Lynch
syndrome (61), and celiac disease (62), where some variants have
enabled prophylactic treatment or screening of other family members,
which may provide cost-effective alternatives to standard disease man-
agement strategies.
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 5
Amidst myriad technological advances and gene discoveries, a
simple family history continues to be advocated as a tool for identification
of common disease risk. For very common conditions with high heri-
tability, such as cardiovascular disease, family history is a much stronger
predictor of disease than any single or combination of genetic/genomic
markers (63). One model suggests that neither family history nor genetic
testing should be used as a standalone but that the real power for disease
prediction, risk assessment, and differential diagnosis comes from their
combined use (63).
Pharmacogenomic markers
GWAS approaches have also been used successfully in pharmaco-
genomics research. Several markers of efficacy, adverse events, and
dosing of therapeutics have been found, but their uptake into clinical
practice is variable, despite the clear actionability of these types of var-
iants. In some cases, such as with the HLA-B*5701 genotype for the
HIV drug abacavir (64)andHLA-B*1502 for the antiseizure drug car-
bamazepine (65), carriers of these genotypes should avoid the drug
entirely to eliminate a specific serious adverse event. In other cases, such
as TPMT for mercaptopurine (66)orCYP2C9/VKORC1 for warfarin
(67,68), adjusting the dose of drug based on genotype can help to avoid
toxicity and improve efficacy. Actionability is not enough to ensure
uptake of pharmacogenomic testing. Such is the case with the anti-
platelet drug clopidogrel, where despite having an FDA black box
warningfor efficacy in individuals carrying the CYP2C19 genetic var-
iant, there is no clear consensus among physicians on its use (69). In
hepatitis C treatment, on the other hand, the IL28B genotype test
not only has proven to be highly predictive of response to pegylated
interferon/ribavirin used to treat chronic HCV infection but also has
seen rapid and widespread adoption in the clinic (70). Genetic markers
that predict reduced therapeutic efficacy may face a high hurdle for
established drugs, unless evidence supporting clinical validity and
utility of the test is indisputable.
Drug repurposing
Among the new programs at the National Center for Advancing Trans-
lational Sciences (http://www.ncats.nih.gov/research/reengineering/
rescue-repurpose/rescue-repurpose.html) is one aimed at using ge-
nomic information to determine whether drugs approved to treat
one disease may be effective in treating others. With 6% of the genome
already being pursued for targets of therapy development (14), addi-
tional investigations into the phenotypes associated with variation in
these targets may open the door to expanded indications. Indeed, two
different researchers recently demonstrated the feasibility of using ge-
nomic information to repurpose drugs. They used computational anal-
ysis to compare gene expression profiles characteristic of certain diseases
with profiles characteristic of specific drugs. By searching for comple-
mentary patterns of gene expression,theysuccessfullyidentifiednew
disease-drug matches, including a match between the antiulcer drug
(cimetidine) and lung cancer (15) and a match between the antiepileptic
drug (topiramate) and inflammatory bowel disease (16), and validated
their predicted use in vivo in rodent models.
Another group used GWAS data for drug repurposing (14). Dis-
ease genes uncovered through GWAS are 2.7-fold enriched for the
molecular targets being pursued by drug developers (14). Besides
the expected overlap in GWAS genes and drug targets for the same
clinical indication (trait), Sanseau and colleagues uncovered more than
100 drug targets that were associated with traits not considered as a
primary indication, opening the door for drug repurposing. In one
example, GWAS identified TNFSF11 variants associated with Crohns
disease. TNFSF11 is the target for the monoclonal antibody denosumab,
currently marketed for osteoporosis, but as a result of this research, it
may also be considered for Crohns disease. Repurposing of existing
FDA-approved drugs eliminates the need for lengthy clinical trials be-
cause formulation, dosing, and safety have already been worked out,
offering new avenues of treatment for these conditions in a short time
frame. Even for drugs in development, knowledge of potential expanded
indications may provide more impetus to push these drugs through de-
velopment. Nonetheless, the practicalities of renavigating the many safe-
ty and efficacy regulatory hurdles should not be underestimated.
Complex multimarker genomic tests for disease diagnosis
and prognosis
Beyond DNA sequence, measures of gene expression, proteins, metab-
olites, and epigenetic changes are being used to generate comprehen-
sive profiles of biological systems in health and disease. Many of the
computational challenges of analyzing these large complex data sets
are being addressed to yield next-generation biomarkers that are mul-
tianalyte, diagnostic, prognostic, and predictive. There are a growing
number of marketed tests that are in vitro diagnostic multivariate
index assays (IVDMIAs), which typically measure protein or RNA
levels, often with complex algorithms, enabling diagnosis and progno-
sis (Table 1) (71). One example is Oncotype Dx (Genomic Health Inc.),
a test that examines expression of 21 genes in tumor tissue to determine
the likelihood of disease recurrence in women with early-stage hormone
estrogen receptorpositive breast cancer (Fig. 2). The test analyzes ex-
pression levels and converts them to a recurrence risk score, which
has been shown to help guide treatment in patients, reduce overall
health care costs, and improve outcomes (7275)andiscurrentlycov-
ered by many major insurance companies. Other examples include
MammaPrint (Agendia Inc.), which analyzes the expression of 70 genes
to determine whether patients are at high or low risk of breast cancer
recurrence, OVA1 (Vermillion Inc.), a five-protein test that gauges
whether a womans ovarian mass is malignant and requires surgery,
AlloMap (XDx Expression Diagnostics Inc.), an 11-gene blood RNA
signature for monitoring rejection after cardiac transplant, and Corus
CAD (CardioDx Inc.), a 23-gene blood RNA signature to screen for
obstructive coronary artery disease.
Despite their complexity, IVDMIAs like these are finding their way
to the market and to the clinic. According to 2007 draft guidance from
the FDA (76), IVDMIAs are being used to make critical health care
decisions and thus should be regulated by the FDA, leading some di-
agnostic developers to work with regulators preemptively. Some, but
not all, of the marketed IVDMIAs have demonstrated analytical and
clinical validity, but evidence of clinical utility is almost always lagging
(77,78). Moreover, the very nature of IVDMIAs presents challenges to
insurers, who grapple with not only limited data on clinical utility but
also how to reimburse such tests that are composed of both a labora-
tory component and an associated algorithm, used to score risk, the
latter part being integral to realizing the testsvalue(79).
The success of some IVDMIAs is a testament not only to the power
of computational biology but also to the importance of advocacy and
financial resources that the commercial developers of these tests bring to
the table. Companies developing IVDMIAs are able to finance pivotal
studies aimed at demonstrating clinical validity, navigate regulatory
hurdles, advocate for coverage by insurance companies, and disseminate
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 6
their tests through marketing to health care providers. Their efforts
offer valuable lessons on the effective translation of genomic tests to
medicine.
Genomics to assess microbial friends and foes
Another exciting area of genomic medicine involves sequencing the
genomes of microorganisms, both the commensal bacteria that reg-
ularly inhabit our bodies (the human microbiome) (80) and the in-
fectious agents that cause acute and sometimes fatal diseases (81).
This past year, the Human Microbiome Project published the first
results of its study of the microbial populations inhabiting various
human body sites (82). Besides providing reference sequences for
many taxa, the major findings from the analysis of this healthy co-
hort included correlation of groups of organisms with host charac-
teristics including ethnicity, age, and body mass index. Further
research in this area has begun to uncover relationships between hu-
man microbial communities and diseases such as diabetes, asthma,
psoriasis, atherosclerosis, obesity, and others (83,84). In one exam-
ple, the gut microbiome has been linked to inflammatory bowel dis-
ease, with patients showing dysbiosis of their gut microbiome,
specifically an expansion of the Proteobacteria phylum that may lead
to inflammation (85). This study, along with others from GWAS
showing that patients with inflammatory bowel disease bear poly-
morphisms of genes conferring gastrointestinal innate immunity,
has led investigators to implicate host immune responses to Proteo-
bacteria in the etiology of inflammatory bowel disease (86). More-
over, strategies to modify the gut microbiome are being explored
as treatments for inflammatory bowel disease, including the use of
fecal microbiota transplantation or engraftment of microbiota from a
healthy donor into a recipient (87). The burgeoning study of the hu-
man microbiome holds tremendous promise for personalized med-
icine because microbial composition can be altered noninvasively
through diet or the use of probiotics or antibiotics.
In the area of infectious disease, NGS of pathogenic microor-
ganisms can supplant the need to first grow them in culture, previ-
ously a major impediment to pathogen identification. For example, in
2003, sequencing of samples from infected patients with the severe
acute respiratory syndrome (SARS) allowed investigators to identify
the causative agent as a coronavirus (88). Comparison of sequences
of multiple isolates of an organism from a single epidemic gives a pic-
ture of the organisms evolution, allowing one to infer where the
outbreak began and how the infection spread. Sequencing was used
to determine the origins of historical outbreaks of cholera (89), tuber-
culosis (90), and the 2009 H1N1 influenza outbreak as well (91). Sim-
ilarly, the source of carbapenem-resistant Klebsiella pneumoniae in a
recent hospital outbreak was identified by sequencing isolates of the
bacteria from infected individuals and examining the genetic dif-
ferences (92). From these data, scientists were able to determine
the chronology of infection, the likely silent carriers who trans-
mitted the infection, and the hospital equipment that may have
acted as a reservoir for the bacteria, even after standard decontami-
nation procedures. The genomic analysis was also able to pinpoint
when resistance to carbapenem first developed. The ability to
sequence microorganisms in a clinically relevant timeframe means
that these methods can now be applied in real time. For example,
an outbreak of methicillin-resistant Staphylococcus aureus was re-
cently curtailed in a hospital after applying genome sequencing to
assess evolution of the epidemic (13).
CHALLENGES AND OPPORTUNITIES FOR GENOMICS
IN MEDICINE
In the past decade, applications of genome research have been pur-
sued across the spectrum of disease management, from risk assess-
ment through diagnosis, prognosis, and treatment. Several themes
in diagnosis and treatment are emerging (Fig. 2). First, as the knowl-
edge of the genomic underpinnings of disease increases, the definition
of self is likewise expanding. This self includes not just the germline
human genome inherited from our parents but also somatic changes
in the genomes of tumors, the genomes of commensal microbes that
regularly inhabit our body, and the genomes of pathogenic organisms.
Second, the influence of genomics on medicine is moving the diagno-
sis of diseases toward the earliest possible point in the course of dis-
ease, before clinical manifestation. Third, genomic testing is occurring
earlier in the life of a human, moving from adulthood to childhood,
the neonatal period, and even prenatally. Finally, pharmacogenomics
is providing a potential means of improving response to therapies, en-
hancing safety, optimizing dosing, and tailoring treatment to the mo-
lecular underpinnings of disease. Notwithstanding this progress,
significant barriers are impeding the rapid translation of genomic dis-
coveries into medical practice.
Whereas research continues on the development, validation, and
utility of genome-based biomarkers, attention is now appropriately
shifting to addressing the roadblocks in translational genomic medi-
cine, from discovery to clinical implementation (93). Here, we highlight
some of the most pressing challenges (Fig. 3) along with innovative so-
lutions. What is becoming clear as the field progresses is that integration
of genomic medicine into practice requires changes in the infrastruc-
ture, processes, and culture along the entire path from discovery to
health care delivery.
Evidentiary framework
Building the evidentiary framework needed to convince the FDA to
approve genomic tests, insurance companies to cover them, and phy-
sicians to use them is perhaps the biggest challenge facing the field of
genomic medicine. In this area, genomic medicine may benefit from
Test awareness
Reimbursement
Regulation
Ethical issues
Physician and
patient education
Clinical
implementation
Evidentiary
framework
Fig. 3. Challenges facing genomic medicine. Shown are the obstacles
on the path from genome discovery to clinical medicine. These obsta-
cles include test awareness (diffusion of innovation), building the evi-
dentiary framework needed to support the clinical validity and utility
of genomic tests, implementation of genomic medicine into the clinical
workflow, ethical issues surrounding genomic testing, regulatory and
reimbursement hurdles, and the education of health care workers and
patients alike.
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 7
partnering with public health agencies, which have vast experience in
epidemiology and surveillance. The Office of Public Health Genomics
at the U.S. Centers for Disease Control and Prevention has developed
a framework for evaluating emerging genetic tests. Their ACCE model
encompasses four areas: analytic validity (how accurately and reliably
the test measures the genotype of interest), clinical validity (how con-
sistently and accurately the test detects or predicts the intermediate or
final outcomes of interest), clinical utility (how likely the test is to sig-
nificantly improve patient outcomes), and ethical, legal, and social im-
plications that may arise in the context of using the test (94). Analytic
validity is usually in the purview of the testing laboratory, which under-
takes a relatively straightforward process to ensure accurate detection
and reporting of the assay results. The bigger challenge is establishing
clinical validity and utility, a process that requires data curation from
the primary scientific literature and, ultimately, carrying out expensive
and time-consuming prospective randomized clinical trials.
The financial burden on a diagnostic developer to conduct random-
ized clinical trials is substantial. As a result, a number of public-
private consortia of stakeholders have emerged to pool resources and
validate genomic biomarkers. The Biomarker Consortium (http://
biomarkersconsortium.org) is one such example where government
and pharmaceutical companies are collaborating on the discovery
and qualification of new biomarkers. The Consortium can leverage
clinical trial data from multiple pharmaceutical companies, which,
when pooled, can markedly increase statistical power and, when
coupled with analytical and scientific expertise from academia, gov-
ernment, and the public sector partners, can accelerate and reduce
the cost of clinical validation.
In the case of rare diseases, which are the focus of clinical sequenc-
ing, or rare outcomes, which are the focus of pharmacogenomic tests
for therapeutic toxicities, enrolling adequate numbers of subjects in a
randomized clinical trial may not be possible. Moreover, randomized
clinical trials do not always represent real-world situations but rather
exemplify an ideal circumstance. To address these concerns, some
have proposed the use of a pragmatic clinical trial design instead
(95,96). In contrast to randomized clinical trials, with their strict pa-
tient eligibility criteria, narrow protocol-driven therapies, and pre-
scribed outcomes, pragmatic clinical trials are conducted in the real-
world medical setting, under best-practice conditions with diverse and
comorbid patient populations. Although less rigorous in design than
randomized clinical trials, the pragmatic clinical trial design could
provide an early indication of the value of pharmacogenomic test-
ing. Comparative effectiveness research is a similarly pragmatic approach
that uses systematic reviews of existing studies, evidence-quality ap-
praisal, and health outcomes research in real-world practice settings
to assess clinical utility. Rapid learning healthcare models are being
used for comparative effectiveness research, whereby diverse and in-
teroperable patient clinical and outcomes data are made available, ide-
ally in a robust and real-time fashion, to potentially support clinical
practice while simultaneously supporting comparative effectiveness
research (97). Coupled with biobanking of patient specimens, the clin-
ical utility of genomic tests can be evaluated in an efficient and cost-
effective manner (98).
Another consideration is that clinical utility conceived only as im-
proved morbidity and mortality fails to appreciate personal utility for
the patient, such as the financial and psychological benefits of resolu-
tion of an unknown diagnosis, changes in life-style leading to overall
improvements in health, or even family planning, all of which are in-
fluenced by knowledge of the genetic susceptibility of disease (99102).
Some groups charged with evaluating genomic tests already recognize
that incorporating the concept of personal utility into assessments of clin-
ical utility will more accurately capture the value of these tests (101).
Through creative partnerships and study designs and a new appreciation
of personal utility, the evidence base for genomic medicine tests will ex-
pand and, in doing so, facilitate approval, coverage, and adoption by the
health care community.
Diffusion of innovation in genomics and medicine
The challenge of diffusion of innovationinhealthcareisnotuniqueto
genomic medicine, but the classical venues for disseminating informa-
tion to health care providers, such as grand rounds and conferences,
may be insufficient for capturing the breadth and depth of genomic
medicine. Passive approaches, including reading the primary scientific
and medical literature, can be time-consuming and overwhelming.
Consequently, several efforts are under way to track and make accessi-
ble the latest available information on genomic tests (Table 2). For drugs
on the market, recommendations for genomic testing can be found in
their labels and in a database of drug-test pairs, currently numbering
more than 100, maintained by the FDA (103). The Pharmacogenomics
Knowledge Base (PharmGKB) is another online resource that includes
information on 186 potentially clinically actionable gene-drug associa-
tions and genotype-phenotype relationships (104). Much of the
information is manually curated from the published literature and is
used to write evidence summaries and pharmacogenomic-based drug
dosing guidelines. Another resource is GeneTests (http://www.ncbi.
nlm.nih.gov/sites/GeneTests), a site focused on Mendelian disorders
that includes a directory of genetic testing laboratories and genetic
and prenatal diagnosis clinics as well as expert-authored peer-reviewed
disease descriptions in a standard format (GeneReviews). GeneTests is
slated to be phased out in 2013 and replaced with the National Institutes
of Healths (NIH) Genetic Testing Registry, a repository for com-
prehensive genetic test information that is voluntarily submitted by
test providers (105). Currently, the site lists 2793 clinical tests, includ-
ing pharmacogenetic and other types of tests that are not in GeneTests.
Perhaps the most comprehensive list of non-Mendelian genomic tests
can be found in a series of Technology Assessment Reports generated
by the Tufts Medical Center Evidence-based Practice Center under
contract to the Agency for Healthcare Research and Quality. Through
horizon scanning of the scientific literature, news, laboratory and com-
mercial Web sites, databases, and other sources, researchers at Tufts
University have populated a database, Gene Test Tracker, with available
clinical genetic tests relevant to the Medicare population. The current
reports cite 154 cancer tests (106) and 127 noncancer genetic tests (107)
in development.
Besides databases for tracking available tests, there is a need for
curation, evaluation, and synthesis of evidence from published re-
search supporting clinical validity and utility of tests to guide clinicians.
Evaluation of Genomic Applications in Practice and Prevention (EGAPP)
(http://www.egappreviews.org/recommendations/index.htm), a group
formedin2004bytheOfficeofPublicHealthGenomics(108), synthesizes
scientific evidence and makes recommendations on appropriate use of
genetic tests in clinical practice. In 2009, PharmGKB and the NIHs
Pharmacogenomics Research Network initiated a similar effort, the
Clinical Pharmacogenetics Implementation Consortium (CPIC) (http://
www.pharmgkb.org/page/cpicGeneDrugPairs) (109). CPIC provides
guidelines to help clinicians understand how available genetic test results
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 8
should be used to optimize drug therapy. Currently, CPIC has published
recommendations for six gene-drug pairs, with eight more under way.
GAPPKB (http://www.hugenavigator.net/GAPPKB/home.do), an in-
tegrated, searchable knowledge base, is also attempting to fill this gap.
This online resource developed by the Office of Public Health Genomics
features the GAPP Finder, a continuously updated, searchable data-
base of genetic tests in transition to practice; Evidence Aggregator,
an application that facilitates searching for evidence reports, systematic
reviews, recommendations, or guidelines in genetic tests and genomic
applications; and PLoS Currents: Evidence on Genomic Tests, an on-
line, open-access journal publishing evidence reviews and recommenda-
tions on genomic tests. These early attempts at supporting diffusion of
innovation are a laudable first step, but a more sustainable means of
curating data on available tests, annotating the scientific literature,
and gathering evidence to support genomic tests in an efficient, dy-
namic, and timely manner needs to be developed.
Clinical implementation
Inorderforgenomicmedicinetobepracticed,itmustbewoveninto
current systems of healthcare delivery, with due consideration not on-
ly to the providers of healthcare but also to the organizations in which
they practice as well. Implementation scientists have outlined various
aspects that need to be considered in order for genomic medicine to
take hold in the clinical setting (110). Beyond the scientific soundness
of the genomic test, measured by a strong evidentiary base and regard
for potential benefits and harms, is consideration of how the new test
will integrate into the clinical workflow. Consideration should be given
to aspects such as access to a laboratory certified by the Clinical Labo-
ratory Improvement Amendments (CLIA) of 1988, methods for sample
preparation and transport, test ordering, receipt, and delivery of results.
Although these features are not unique to genomic testing, their imple-
mentation is complicated by issues of privacy, complex interpretation of
results, and the need to involve third parties for counseling in some
cases. These facets may require the development of new systems to ac-
commodate genomic tests (111). For example, surgical and interven-
tional radiology services are accustomed to fixing tissues in formalin
to preserve them for analysis at a later date, but NGS works best on
fresh tissues. Either adapting NGS technology to work within the con-
straints of current tissue handling procedures or modifying those pro-
cedures to allow collection of fresh or fresh-frozen tissues may be
necessary (112).
A robust means of integrating genomic data into electronic health
records will be required, with consideration of not only data storage
formats and privacy issues but also appropriate decision support tools
for prompting their use at the point of care and delivering results in an
easily interpretable format (113115). To meet these needs, NGS
companies like Knome (http://www.knome.com) are developing their
own plug-and-play bioinformatic support tools, and commercial ven-
dors of electronic health records have begun to address genomic-
related applications as well (113). Some patients may opt to self-manage
their own genomic data in a personal health record such as Microsoft
Health Vault (http://www.microsoft.com/en-us/healthvault) or Dossia
Table 2. Genomic medicines address book. Shown are sites that curate available genomic tests and the evidence to support their use.
Web site URL Description
CPIC http://www.pharmgkb.org/page/cpic Provides freely available, peer-reviewed, updatable, and detailed gene/drug
clinical practice guidelines; 6 currently published, 8 under way
EGAPP http://www.egappreviews.org/ Synthesizes scientific evidence and makes recommendations on appropriate
use of genetic tests in clinical practice; 8 evidence reports and 6
recommendations currently available
Evidence aggregator http://www.hugenavigator.net/GAPPKB/
evidencerStartPage.do
Search engine for evidence reports, systematic reviews, recommendations, or
guidelines in genetic tests and genomic applications
FDA biomarkers http://www.fda.gov/drugs/scienceresearch/
researchareas/pharmacogenetics/
ucm083378.htm
List of pharmacogenomic biomarkers on drug labels (link to
drug labels provided); currently includes >100 biomarker-drug pairs
GAPP Finder http://www.hugenavigator.net/GAPPKB/
topicStartPage.do
A continuously updated, searchable database of genetic tests in transition to
practice; currently includes 519 tests
GeneTests http://www.ncbi.nlm.nih.gov/sites/GeneTests/ Information on genetic tests, mostly for rare Mendelian disorders. Site
includes directory of testing laboratories, genetic and prenatal diagnosis
clinics, and expert-authored peer-reviewed disease descriptions
(GeneReviews); currently includes >2900 disease entries; due to be phased
out in 2013
Genetic testing registry http://www.ncbi.nlm.nih.gov/gtr/ Central location for voluntary submission of genetic test information by
providers; includes information on test methodology, validity, evidence
of the tests usefulness, and laboratory contacts and credentials;
currently includes >1200 fully registered tests for >500 conditions
PharmGKB http://www.pharmgkb.org Information on potentially clinically actionable gene-drug associations
and genotype-phenotype relationships; currently lists 186 well-known
pharmacogenomic associations and provides 46 summaries for very
important genes
PLoS currents: evidence
on genomic tests
http://currents.plos.org/genomictests/ Online, open-access journal publishing evidence reviews and recommendations
on genomic tests; currently has 23 genomic test reviews
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 9
(http://www.dossia.org) as a safeguard. Currently, there are several
examples of decision support tools, such as Warfarin Dosing (http://
www.WarfarinDosing.org), but they are typically standalone tools and
not part of routine clinical workflow. To maximize their effectiveness,
such tools should be integrated into electronic health records. Tapping
into the collective knowledge and experience of various institutions
working in this space would greatly facilitate this effort. Ultimately, a na-
tional, standardized technical architecture for integrating clinical decision
support into electronic health records will be required. Notable efforts in
this space include those of Health Level 7 (http://www.hl7.org), an orga-
nization that provides interoperability standards for the exchange, integra-
tion, sharing, and retrieval of electronic health information. Through their
Clinical Genomics Workgroup, this organization has developed a stan-
dards guide for genetic testing that includes document templates to
support integration of genetic testing into electronic health records
(116). Appropriate clinical decision support, provided in the context of
the electronic health record, will greatly facilitate the diffusion and uptake
of genomic medicine.
Regulation of genomic tests
Regulationofgenomictestsisbothapublic health issue and, ultimate-
ly, an economic one. Physicians and patients alike look to government
regulators to assure them that the tests have been carefully scrutinized
for their safety, efficacy, and intrinsic value. Currently, two federal or-
ganizations are in charge of regulating genetic tests (117). A small per-
centage of genetic tests are sold as diagnostic devices, meaning that a
company makes and sells genetic test kits to a laboratory for testing,
and these are regulated by the FDAs Office of In Vitro Diagnostics.
Before marketing, the analytical validity of the device must be assessed,
but in cases where clinical performance has not been well established,
the clinical validity is examined as well. The specific degree of regula-
tion of medical devices is currently tailored to their level of risk: class I
(low risk, few regulatory controls), class II (moderate risk), and class
III (high risk, more controls, including submission of a premarket ap-
proval application) (117). Many genomic tests will fall into class II, as
is the case with genetic tests for drug-metabolizing enzymes (118).
Some genomic tests will fall into class III, for example, those that alter
a therapeutic decision in such a way as to expose individuals to po-
tentially harmful treatments, such as radiation therapy in cancer. Re-
cent guidance from the FDA clarifies the principal factors it considers
when making benefit-risk determinations on medical devices (119).
Most genetic tests today are developed and offered by individual
laboratories as laboratory-developed tests, and these laboratories are
overseen by the Centers for Medicare and Medicaid Services (CMS).
CMS is primarily concerned with monitoring the laboratoryscompli-
ance with CLIA regulations in their testing procedures. For years, the
FDA has claimed enforcement discretion and opted not to regulate
laboratory-developed tests unless they were deemed high risk,as
in the case of IVDMIAs (76). However, the selective approach to reg-
ulate only a subset of high-risk laboratory-developed tests has led to
more confusion and created an uneven playing field. Moreover, the
rise of direct-to-consumer genetic testing companies, those that bypass
physicians and market their services directly to patients outside the med-
ical setting, has brought regulatory oversight of genomic tests in general
to the forefront (120). On the basis of a series of recommendations
by the U.S. Department of Health and Human Services Secretarys
Advisory Committee on Genomics, Health, and Society to enhance
oversight (121), the FDA has responded with a 2010 notice stating that
they may actively regulate not just high-risk tests but all laboratory-
developed tests (122). Regulation of genomic tests is a moving target
that needs to strike a balance between commercial interests and pa-
tient safety to move genomic medicine forward.
Coverage and reimbursement
Although FDA approval of a genomic medicine test may improve the
likelihood of coverage by insurance companies, it is no guarantee.
Whether an insurance plan covers genetic testing depends on several
factors, including consumer demand, opinions of professional organi-
zations, integration into clinical guidelines, and, most importantly, the
strength of evidence supporting a tests analytical and clinical validity
and clinical utility (123). Private insurance plans all make their own
decisions regarding whether to cover and how much to reimburse for
a genomic test, and it can vary on a case-by-case basis within those
plans. Nonetheless, private insurers often mimic the coverage decisions
of Medicare, as the largest provider of health insurance in the United
States. Medicare decisions are made by CMS, which has a policy of
reimbursing tests that are reasonable and necessary for diagnosis or
treatment of an illness or injury. However, CMS does not typically re-
imburse screening tests, including genetic predisposition tests, except
in the presence of signs and symptoms of disease. CMSs Medical
Evidence Development and Coverage Advisory Committee met in 2009
to discuss what types of evidence will be needed to evaluate screen-
ing genetic tests for Medicare coverage (124). This information should
helpguidefuturedevelopmentofgenomicteststoimprovetheirlike-
lihood of coverage.
In a further sign that the field is moving forward, the American
Medical Association issued a new coding system for molecular diag-
nostics, and the CMS has been deliberating on how to reimburse these
tests (125).Oneofthekeychallengeswillbeobtainingreimbursement
not just for the pathology laboratories that are running the tests but
also for the physicians or other health care workers who are interpret-
ing the tests. Coverage and reimbursement of genomic tests is evolving,
but in the current system of healthcare delivery, it remains an essential
component that needs to be addressed to see widespread uptake of ge-
nomic medicine.
Ethical, legal, and social issues
Many of the ethical, legal, and social issues related to genomic medicine
such as patient privacy, selective termination of pregnancies based on
genetic information, and patentability of DNAhave existed since the
arrival of genetic testing. In a current landmark case sparked by con-
troversy over the BRCA1 gene patent held by Myriad Genetics
Association for Molecular Pathology, et al. v. U.S. Patent and Trade-
mark Office, et al.the U.S. Supreme Court is hearing arguments on
the patentability of genes (126). The ruling will have implications for
diagnostic use of gene patents, affecting developers of diagnostics and
patients alike. Privacy is another issue that has been reignited by the
introduction of NGS and fueled by an atmosphere of collaborative,
open-access public data sharing. Studies of publicly available sequence
data have shown that patients in research studies can be identified by
their genome sequences (127,128). These findings demonstrate the
potential for sequence data to expose patient identities, with significant
implications for not only the individual but also their family members
and possibly the larger community to which they belong. The 2008
Genetic Information Nondiscrimination Act is a U.S. federal law
that bars insurers and employers from discriminating on the basis
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 10
of genetic information, but it does not pertain to long-term disability
or life insurance, nor does it protect against stigmatization. The
application of NGS to noninvasive prenatal testing has also rekindled
an old concern over the misuse of genetic information to perform se-
lective abortion, already an ethically and politically contentious issue.
Fear that decisions will be made on the basis of trivial genetic traits has
reopened the debate about which health conditions, if any, are suffi-
cient grounds to justify abortion (129).
The ability to uncover all genetic variants in a subject through
NGS also raises a new dilemma: whether to disclose to patients poten-
tially clinically meaningful variants unrelated to the primary indication
for sequencing, so-called secondary or incidental findings (128,130).
On average, every human has several hundred coding region variants
in their genomes, most of which have unknown health consequences
(6). Who decides which of the discovered mutations are clinically rel-
evant and under what circumstances the incidental findings of NGS,
of either known or unknown health significance, should be shared
with patients are hotly debated topics. The American College of Med-
ical Genetics advocates for clear policies related to return of results,
stating that patients should be informed of those policies and under-
stand what types of information might be reported back to them and
under what circumstances, as well as recommending that patients
should be able to opt out of receiving certain findings (131). In their
recently published position statement, the American College of Med-
ical Genetics recommends that laboratories conducting exome and ge-
nome sequencing for clinical use should notify physicians about their
patientsstatus for a number of specific conditions, genes, and variants
that are found during the sequencing (132). This position has been
controversial because it is seen to be at odds with patient autonomy
and the rights of children to not learn about genetic predisposition to
adult-onset conditions. This has led the American College of Medical
Genetics to release a statement clarifying their position (133).
The limited understanding of the health implications of genetic
variation uncovered through NGS is concerning. In 2011, the National
Human Genome Research Institute sponsored a workshop, ClinAction,
to devise a plan for systematically evaluating and cataloguing genetic
variants based on their clinical actionability (134). The experts in attend-
ance proposed storing data on actionable variants in a new database,
ClinVar (http://www.ncbi.nlm.nih.gov/clinvar), which could be integrated
into the clinical workflow. Initially, the database will be populated with
existing variants catalogued in the Online Mendelian Inheritance in Man
database (http://www.omim.org), GeneReviews (http://www.ncbi.nlm.nih.
gov/sites/GeneTests/review), Locus-Specific Mutation Databases (http://
www.hgvs.org/dblist/glsdb.html), and other sources, but will grow as
new discoveries are made. An important aspect of this effort is the need
to guard against false-positive results that can occur in NGS studies based
on single patients or families, and for which very strict peer review of the
scientific evidence linking the genetic variant to disease is warranted. For-
tunately, discourse on incidental genetic findings as well as other ethical
issues is occurring in parallel with development of genomic medicine as
the research community strives to balance innovation with responsible
use of new technologies.
Education
There is a growing sentiment that uptake of genomic medicine is slow
becausehealthcareproviderslackadequate training in genomics. One
study found that describing a test as genetic (versus nongenetic) sig-
nificantly decreased a physicians likelihood of offering the test (135).
Primary healthcare providers will likely assume prominent roles at the
front lines of genomic medicine, taking responsibility for administer-
ing new tests and fielding questions from informed patients. Hence,
they will not only need to be armed with practical information on
what tests are available, when and how to use them, where to get them
done, and what to tell patients, but also require a conceptual founda-
tion on which to build their capacitytoevaluateanddelivergenomics
in the course of clinical care. Although most physicians have heard of
pharmacogenomics (136), few are aware of direct-to-consumer genetic
testing (137), and with the rapid pace of discovery, staying on top
of the latest available genomic medicine tools will be a daunting task. Re-
search has shown that few primary care providers feel comfortable or-
dering genomic tests or explaining test results to patients (136,137),
and most feel an urgent need for genetics education (138).
In their report on genetics education, the U.S. Department of
Health and Human Services Secretarys Advisory Committee on Ge-
nomics, Health, and Society highlighted factors that contribute to the
limited genetics education of healthcare professionals, including issues
such as a crowded curricula, lack of knowledgeable faculty, lack of
evidence-based guidelines in genetics, and misconceptions about the
nature of genomic medicine (139). The committee recommended
modifications in medical, dental, nursing, public health, and pharmacy
school curricula and in medical residencytrainingprogramstoensure
that healthcare professionals entering the workforce are well trained in
genetics. These recommendations prompted the National Coalition
for Health Professional Education in Genetics to develop specific core
competencies for all health care professionals on knowledge, skills, and
attitudes needed to effectively deliver genomic medicine (140).
But still unresolved are the best methods to educate health care
providers. In the last few years, several university medical schools have
made significant efforts to fill the genomics education gap (141144).
These efforts include targeting curriculum to medical students and res-
idents in training and continuing medical education for practicing
professionals. There is a push to introduce topics related to genomic
medicine even earlier, during undergraduate training. However, improv-
ing overall genomics literacy of health care providers is only one piece
of the puzzle, and keeping pace with this rapidly evolving field will be
a continuous challenge.
Enhanced genomic education not only is key for health care pro-
viders but also extends to all professions that genomic medicine touches
upon. Policy-makers, regulators, lawyers, investors, insurance under-
writers, and others will need some understanding of genomics to
move genomic medicine forward. It remains unclear how these pro-
fessionals will receive this information. Moreover, acceptance of ge-
nomics ultimately may require a level of comfort on the part of the
patient, who may be concerned about privacy and discrimination or
confused about the implications of a particular genetic test result. This
may be especially problematic for patients obtaining genomic data
through direct-to-consumer test providers. Studies find that more
than one-third of U.S. adults have limited health literacy (145), which
in turn affects understanding of print and oral communications about
genetic and genomic information (146). Historically, genetic counsel-
ors have been responsible for putting genetic information into context
for patients undergoing genetic testing, but these situations have
mainly involved Mendelian traits with clear inheritance and strong
effects and have occurred in the domain of specialty genetics clinics.
As genomics spreads from specialty clinics to more mainstream medi-
cine where primary care physicians will be utilizing genomic information
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 11
for clinical decision-making, the demand for genetic counselors may
far outstrip the supply, making this model impractical. Alternative so-
lutions, including additional training, and perhaps certification, of
primary health care providers in the delivery of genomic medicine will
need to be considered. In addition, improvement in the formal primary
and secondary education of consumers in the areas of scientific and
health literacy will enhance informed decision-making (93).
FUTURE EXPECTATIONS
The late 1990s marked what many consider to be the dawn of the era
of personalized medicine, when the full human genome was being re-
vealed for the first time and scientists were beginning to explore the
many ways that this information could be used to improve medicine
(147). Today, genome research has delivered promising new tools for
disease management, and early signs of uptake at academic medical
centers are encouraging. To be sure, the pace of implementation has
been slower than anticipated, and genomic medicine is far from
mainstream. Nonetheless, the challenges that prevent more widespread
uptake are not intractable but will require a concerted effort by the
many stakeholders in the field to overcome.
Moving forward, technological advances will continue to drive
discovery. New applications of NGS are on the horizon, including
profiling of immune cell repertoires (5) for monitoring hematological
malignancies (148,149) and responses to vaccines (150), and detecting
solid transplant rejection (151), just to name a few. Third-generation
sequencing platforms are expected to provide longer read lengths,
chromosome phasing, and reduced time and cost, increasing the ac-
curacy, capacity, and turnaround time for genome sequencing (152).
Not only will future research efforts lead to new markers and tools to
predict and manage disease at an increasing rate, but we also may see
fundamental shifts in how we define disease, how medicine is de-
livered to patients, and how consumers manage their own health
and affect change.
Creative partnerships and a rise in consumer-driven
genomic research
Thepaceofgenomicbiomarkerdiscovery will continue to accelerate
through creative research and funding partnerships. Large disease-
centered consortia are already bringing together investigators to col-
laborate and pool data and resources. For example, the International
Cancer Genome Consortium (153) involves 51 project teams contribut-
ing genomic data from 24,000 tumors across a wide spectrum of
cancers (http://icgc.org). The open-access movement will continue to
promote data sharing through portals such as Gene Expression Omnibus
(http://www.ncbi.nlm.nih.gov/geo) and dbGaP (http://www.ncbi.nlm.
nih.gov/gap), where any qualified investigator can generate hypotheses
and analyze data. Educated patients are also becoming a driving force
behind personalized medicine. The movement toward consumer-driven
health care has empowered patients, leading them to seek information
on medical conditions through the Internet, track their own health
history through personal health records, and obtain genomic data
through direct-to-consumer genetic testing companies. The downside
of this trend is that, without a medical professional to help with the inter-
pretation of the information, patients may misinterpret their genetic in-
formation, which could lead to unnecessary stress or misinformed health
decisions. Patients are now networked through social media, sharing
information on health conditions through sites like PatientsLikeMe
(http://www.patientslikeme.com) and, in some cases, bypassing the
medical establishment altogether. Crowd-sourcing, crowd-funding,
and participant-driven research are also on the rise. The direct-to-
consumer genetics company 23andMe has proven the feasibility of
web-based collection of self-reported data from an engaged cohort
of research participants that can beusedforgenomicresearchinsome
cases (154). Although these efforts are not without their caveats, in-
cluding the potential for bias and confounding leading to spurious
results (155,156), and will not replace carefully designed epidemiological
studies, they represent an innovative approach to overcoming some of
the financial and logistical constraints inherent in genomic research.
Another example of this trend is the newly launched American Gut
project (http://humanfoodproject.com/american-gut), an open-source,
community-driven effort to characterize the microbial diversity of the
American public where study subjects are recruited online and not
only participate in research but also help to fund it.
New taxonomy for disease
In the coming years, we will likely see a fundamental shift in how
disease is classified. Currently, diseases are classified on the basis of
subjective clinical signs and symptoms and, in some cases, objective
laboratory or image-based tests. Absent from this framework in
most cases is a measure of the perturbed molecular pathways that
characterize the disease. Genomics is poised to deliver molecular-
level definitions of the physiological processes underlying diseases,
suggesting treatments targeted at the molecular lesions instead of
thesymptoms.Nowhereisthismoreevidentthanincancer,where
subclassification of disease is having a tangible effect on treatment
and where tissue of origin is secondary to the molecular profile of the
tumor. The current disease classification system used in the health care
industry as a basis for diagnosis and reimbursement is the International
Statistical Classification of Diseases and Related Health Problems (ICD),
currently in its 10th revision (ICD-10) (157). The ICD-10 for oncology
already incorporates genomic factors to distinguish subtypes of can-
cer (158160). ICD-11, the next revision, due to be published in 2015,
promises to expand this theme. Momentum on creating a new disease
taxonomy is growing, as evidenced by an extensive report on devel-
opment of precision medicine by the National Research Council of the
U.S. National Academies of Science (161) and the prioritization of
precision and personalized medicine this year by the World Economic
Forum (162).
New methods for delivering genomic medicine preemptively
Because the cost of sequencing continues to decline and the analyt-
ical issues abate, it is conceivable that individuals will have their ge-
nome sequenced at some point in their lives, perhaps even at birth.
Health systems will likely incorporate all or part of ones genomic in-
formation into their electronic medical record. Thus, critical informa-
tion on pharmacogenetic markers of toxicity or drug response, for
example, would be available preemptively, before prescribing a drug.
This model is already being piloted in the NIHseMERGEprogram
(163) at Vanderbilt University as part of their PREDICT initiative
(164) and at other institutions as well (165). Compared to current test-
ing practices, such an approach could provide a more cost-effective
means to capture and treat rare, preventable genetic diseases missed
by the current health care system, aid in future family planning, and
improve safety and efficacy of therapies.
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 12
Point-of-care diagnostics and digital medicine
New ways of delivering personalized medicine are being developed for
use at the bedside and beyond. Point-of-care diagnostic tests will facil-
itate rapid diagnosis at the patients bedside, physiciansoffices, emer-
gency rooms, and at home (166). We will see a rise in the use of digital
medicine, where patients monitor their own vital statistics at home
through sensors and handheld devices and send data directly to their
doctor (167). One day, this monitoring may include dynamic measures
of the genome including longitudinal, integrated personal omicspro-
files (168)orgeneexpressionchangesindicativeofexposuretoinfec-
tiousagents(169).
Rise in third-party genomic information brokers
Genome interpretive services will emerge to assist clinicians in under-
standing the meaning and actionability of genome information, much
the same way as radiologists assist in the interpretation of imaging.
This nascent field currently includes startup companies like Knome
(http://www.knome.com), Personalis (http://www.personalis.com),
Omicia (http://www.omicia.com), Genomatix (http://www.genomatix.
de), Cypher Genomics (http://cyphergenomics.com), Silicon Valley
Biosystems (http://www.svbio.com), and GenomeQuest (http://www.
genomequest.com), who are offering software, computer infrastruc-
ture, and services required to process, analyze, and store patient sequence
data and, in some cases, even produce tailored diagnostic reports.
CONCLUSION
Genomic medicineonly an aspiration 10 years agois beginning
to emerge across the entire clinical continuum from risk assessment in
healthy individuals to genome-guided treatment of complex diseases
in patients. Technology continues to propel the field forward, but
translating discovery into routine use is complex, requiring changes
in the fundamental processes of regulation, reimbursement, and clin-
ical practice. Progress is tempered by consideration of ethical issues
and the need to fill the education gap that exists for health care pro-
viders and consumers alike, which makes it difficult to keep pace with
advances in the field. External forces from social networking to digital
and information technologies are enabling consumers to take health
matters into their own hands, generating momentum like never before.
Today, we are on a path toward implementation of genomic medicine,
but that path is long, mired with obstacles, and potentially perilous.
Moreover, it remains to be seen whether genomic medicine will actually
improve health, when efforts to implement simpler clinical and preven-
tive strategies have failed. Nonetheless, the movement toward improv-
ing disease diagnosis and treatment with genomics is unlikely to halt
and there is reason to be optimistic. How will we know when we have
arrived? When we stop talking about genomic medicine, and refer to it
simply as medicine.
REFERENCES
1. B. B. Spear, M. Heath- Chiozzi, J. Huff, Clinical applic ation of pharmacogenetics. Trends
Mol. Med. 7, 201204 (2001).
2. R . J. Klein, C. Zeiss, E. Y. Chew, J. Y. Tsai, R. S. Sackler, C. Haynes, A. K. Henning, J. P. SanGiovanni,
S. M. Mane, S. T. Mayne, M. B. Bracken, F. L. Ferris , J. Ott, C. Barnstable, J. Hoh , Com-
plement factor H polymorphism in age-related macular degeneration. Science 308,
385389 (2005).
3. K. A. Wetterstrand, DNA sequencing costs: Data from the NHGRI Genome Sequencing
Program (GSP); http://www.genome.gov/sequencingcosts.
4. E. S. Lander, Initial impact of the sequencing of the human genome. Nature 470, 187197
(2011).
5. S. D. Boyd, Diagnostic applications of high-throughput DNA sequencing. Annu. Rev.
Pathol. 8, 381410 (2013).
6. 1000 Genomes Project Consortium, G. R. Abecasis, A. Auton, L. D. Brooks, M. A. DePristo,
R. M. Durbin, R. E. Handsaker, H. M. Kang, G. T. Marth, G. A. McVean, An integrated map of
genetic variation from 1,092 human genomes. Nature 491,5665 (2012).
7. M.B.Gerstein,A.Kundaje,M.Hariharan,S.G.Landt,K.K.Yan,C.Cheng,X.J.Mu,E.Khurana,
J.Rozowsky,R.Alexander,R.Min,P.Alves,A.Abyzov,N.Addleman,N.Bhardwaj,A.P.Boyle,
P. Cayting, A. Charos, D. Z. Chen, Y. Cheng, D. Clarke, C. Eastman, G. Euskirchen, S. Frietze,
Y.Fu,J.Gertz,F.Grubert,A.Harmanci,P.Jain,M.Kasowski,P.Lacroute,J.Leng,J.Lian,
H. Monahan, H. OGeen, Z. Ouyang, E. C. Partridge, D. Patacsil, F. Pauli, D. Raha, L. Ramirez,
T. E. Reddy, B. Reed, M. Shi, T. Slifer, J.Wang, L. Wu, X. Yang, K. Y. Yip, G. Zilberman-Schapira,
S. Batzoglou, A. Sidow, P. J. Farnham, R. M. Myers, S. M. Weissman, M. Snyder, Architecture
of the human regulatory network derived from ENCODE data. Nature 489,91100 (2012).
8. S. B. Ng, E. H. Turner, P. D. Robertson, S. D. Flygare, A. W. Bigham, C. Lee, T. Shaffer, M. Wong,
A. Bhattacharjee, E. E. Eichler, M. Bamshad, D. A. Nickerson, J. Shendure, Targeted capture
and massively parallel sequencing of 12 human exomes. Natu re 461,272276 (2009).
9. S.B.Ng,K.J.Buckingham,C.Lee,A.W.Bigham,H.K.Tabor,K.M.Dent,C.D.Huff,P.T.Shannon,
E. W. Jabs, D. A. Nickerson, J. Shendure, M. J. Bamshad, Exome sequencing identifies the
cause of a mendelian disorder. Nat. Genet. 42,3035 (2010).
10. M. Choi, U. I. Scholl, W. Ji, T. Liu, I. R. Tikhonova, P. Zumbo, A. Nayir, A. Bakkaloğlu, S. Ozen,
S. Sanjad, C. Nelson-Williams, A. Farhi, S. Mane, R. P. Lifton, Genetic diagnosis by whole
exome capture and massively parallel DNA sequencing. Proc. Natl. Acad. Sci. U.S.A. 106,
1909619101 (2009).
11. B. Rabbani, N. Mahdieh, K. Hosomichi, H. Nakaoka, I. Inoue, Next-generation sequencing:
Impact of exome sequencing in characterizing Mendelian disorders. J. Hum. Genet. 57,
621632 (2012).
12. F. S. Ong, K. Das, J. Wang, H. Vakil, J. Z. Kuo, W. L. Blackwell, S. W. Lim, M. O. Goodarzi,
K.E.Bernstein,J.I.Rotter,W.W.Grody,Personalizedmedicineandpharmacogenetic
biomarkers: Progress in molecular oncology testing. Expert Rev. Mol. Diagn. 12,593602
(2012).
13. S.R.Harris,E.J.Cartwright,M.E.Török,M.T.Holden,N.M.Brown,A.L.Ogilvy-Stuart,
M. J. Ellington, M. A. Quail, S. D. Bentley, J. Parkhill, S. J. Peacock, Whole-genome sequenc-
ing for analysis of an outbreak of meticillin-resistant Staphylococcus aureus: A descriptive
study. Lancet Infect. Dis. 13,130136 (2013).
14. P. Sanseau, P. Agarwal, M. R. Barnes, T. Pastinen, J. B. Richards, L. R. Cardon, V. Mooser,
Use of genome-wide association studies for drug repositioning. Nat. Biotechnol. 30,317320
(2012).
15. M. Sirota, J. T. Dudley, J. Kim, A. P. Chiang, A. A. Morgan, A. Sweet-Cordero, J. Sage, A. J. Butte,
Discovery and preclinical validation of drug indications using compendia of public gene
expression data. Sci. Transl. Med. 3, 96ra77 (2011).
16. J.T.Dudley,M.Sirota,M.Shenoy,R.K.Pai,S.Roedder,A.P.Chiang,A.A.Morgan,M.M.Sarwal,
P. J. Pasricha, A. J. Butte, Computational repositioning of the anticonvulsant topiramate for
inflammatory bowel disease. Sci. Transl. Med. 3, 96ra76 (2011).
17. D. J. Slamon, G. M. Clark, S. G. Wong, W. J. Levin, A. Ullrich, W. L. McGuire, Human breast
cancer: Correlation of relapse and survival with amplification of the HER-2/neu oncogene.
Science 235, 177182 (1987).
18. D. J. Slamon, B. Leyland-Jones, S. Shak, H. Fuchs, V. Paton, A. Bajamonde , T. Fleming,
W. Eiermann, J. Wolter, M. Pegram, J. Baselga, L. Norton, Use of chemotherapy plus a
monoclonal antibody against HER2 for metastatic breast cancer that overexpresses
HER2. N. Engl. J. Med. 344, 783792 (2001).
19. S. J. Schnitt, T. W. Jacobs, Current status of HER2 testing: Caught between a rock and a
hard place. Am. J. Clin. Pathol. 116, 806810 (2001).
20. M. E. Hammond, P. L. Fitzgibbons, C. C. Compton, D. J. Grignon, D. L. Page, L. P. Fielding,
D. Bostwick, T. F. Pajak, College of American Pathologists Conference XXXV: Solid tumor
prognostic factorsWhich, how and so what? Summary document and recommenda-
tions for implementation. Cancer Committee and Conference Participants. Arch. Pathol.
Lab. Med. 124, 958965 (2000).
21. V. Pawlowski, F. Révillion, L. Hornez, J. P. Peyrat, A real-time one-step reverse transcriptase-
polymerase chain reaction method to quantify c-erbB-2 expression in human breast cancer.
Cancer Detect. Prev. 24, 212223 (2000).
22. J. S. Ross, E. A. Slodkowska, W. F. Symmans, L. Pusztai, P. M. Ravdin, G. N. Hortobagyi, The
HER-2 receptor and breast cancer: Ten years of targeted antiHER-2 therapy and person-
alized medicine. Oncologist 14, 320368 (2009).
23. T.J.Lynch,D.W.Bell,R.Sordella,S.Gurubhagavatula,R.A.Okimoto,B.W.Brannigan,
P.L.Harris,S.M.Haserlat,J.G.Supko,F.G.Haluska,D.N.Louis,D.C.Christiani,J.Settleman,
D. A. Haber, Activating mutations in the epidermal growth factor receptor underlying respon-
siveness of nonsmall-cell lung cancer to gefitinib. N. Engl. J. Med. 350,21292139 (2004).
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 13
24. J.G.Paez,P.A.Jänne,J.C.Lee,S.Tracy,H.Greulich,S.Gabriel,P.Herman,F.J.Kaye,
N. Lindeman, T. J. Boggon, K. Naoki, H. Sasaki, Y. Fujii, M. J. Eck, W. R. Sellers, B. E. Johnson,
M. Meyerson, EGFR mutations in lung cancer: Correlation with clinical response to gefitinib
therapy. Science 304,14971500 (2004).
25. C.J.Allegra,J.M.Jessup,M.R.Somerfield,S.R.Hamilton,E.H.Hammond,D.F.Hayes,
P. K. McAllister, R. F. Morton, R. L. Schilsky, American Society of Clinical Oncology
pro visional clinical opinion: Testing for KR AS gene mutations in patients with metastatic co-
lorectal carcinoma to predict response to antiepidermal growth factor receptor monoclonal
antibody therapy. J. Clin. Oncol. 27, 20912096 (2009).
26. U.S. Food and Drug Administration, Cetuximab in combination with Folfiri/Therascreen;
http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm310933.htm.
27. U.S. Fo od and Drug Administration, Drug app roval package: Xalkori capsule s; http://
www.accessdata.fda.gov/drugsatfda_docs/nda/2011/202570Orig1s000TOC.cfm.
28. U .S. Food and Drug Administration, Drug approval package: Zelboraf (vemurafenib)
tablet; http://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/202429s000TOC.cfm.
29. M. Röring, T. Brummer, Aberrant B-Raf signaling in human cancer10 years from bench
to bedside. Crit. Rev. Oncog. 17,97121 (2012).
30. E. Tiacci, V. Trifonov, G. Schiavoni, A. Holmes, W. Kern, M. P. Martelli, A. Pucciarini, B. Bigerna,
R. Pacini, V. A. Wells, P. Sportoletti, V. Pettirossi, R. Mannucci, O. Elliott, A. Liso, A. Ambrosetti,
A. Pulsoni, F. Forconi, L. Trentin, G. Semenzato, G. Inghirami, M. Capponi, F. Di Raimondo,
C. Patti, L. Arcaini, P. Musto, S. Pileri, C. Haferlach, S. Schnittger, G. Pizzolo, R. Foà, L. Farinelli,
T. Haferlach, L. Pas qualucci, R. Ra badan, B. Falini , BRAF mutations in hai ry-cell leukem ia.
N. Engl. J. Med. 364, 23052315 (2011).
31. S. Dietrich, H. Glimm, M. Andrulis, C. von Kalle, A. D. Ho, T. Zenz, BRAF inhibition in refrac-
tory hairy-cell leukemia. N. Engl. J. Med. 366, 20382040 (2012).
32. Y.P.Mosse,F.M.Balis,M.S.Lim,J.Laliberte,S.D.Voss,E.Fox,R.Bagatell,B.Weigel,
P. C. Adamson, A. M Ingle, C. H. Ahern, S. Blaney, Efficacy of crizotinib in children with
relapsed/refractory ALK-driven tumors including anaplastic large cell lymphoma and
neuroblastoma: A Childrens Oncology Group phase I consortium study. J. Clin. Oncol.
30, abstr. 9500 (2012).
33. G. Iyer, A. J. Hanrahan, M. I. Milowsky, H. Al-Ahmadie, S. N. Scott, M. Janakiraman, M. Pirun,
C.Sander,N.D.Socci,I.Ostrovnaya,A.Viale,A.Heguy,L.Peng,T.A.Chan,B.Bochner,
D. F. Bajorin, M. F. Berger, B. S. Taylor, D. B. Solit, Genome sequencing identifies a basis
for everolimus sensitivity. Science 338, 221 (2012).
34. R.Katayama,A.T.Shaw,T.M.Khan,M.Mino-Kenudson,B.J.Solomon,B.Halmos,N.A.Jessop,
J.C.Wain,A.T.Yeo,C.Benes,L.Drew,J.C.Saeh,K.Crosby,L.V.Sequist,A.J.Iafrate,J.A.Engelman,
Mechanisms of acquired crizotinib resistance in ALK-rearranged lung cancers. Sci. Transl. Med.
4, 120ra17 (2012).
35. H. L. McLeod, Cancer pharmacogenomics: Early promise, but concerted effort needed.
Science 339, 15631566 (2013).
36. R.J.Leary,I.Kinde,F.Diehl,K.Schmidt,C.Clouser,C.Duncan,A.Antipova,C.Lee,K.McKernan,
F. M. De La Vega, K. W. Kinzler, B. Vogelstein, L. A. Diaz Jr., V. E. Velculescu, Development of
personalized tumor biomarkers using massively parallel sequencing. Sci. Transl. Med. 2,
20ra14 (2010).
37. R. J. Leary, M. Sausen, I. Kinde, N. Papadopoulos, J. D. Carpten, D. Craig, J. OShaughnessy,
K. W. Kinzler, G. Parmigiani, B. Vogelstein, L. A. Diaz Jr., V. E. Velculescu, De tection of
chromosomal alterations in the circulation of cance r patients with whole-genome se-
quencing. Sci. Transl. Med. 4, 162ra154 (2012).
38. S. J. Dawson, D. W. Tsui, M. Murtaza, H. Biggs, O. M. Rueda, S. F. Chin, M. J. Dunning, D. Gale,
T. Forshew, B. Mahler-Araujo, S. Rajan, S. Humphray, J. Becq, D. Halsall, M. Wallis, D. Bentley,
C. Caldas, N. Rosenfeld, Analysis of circulating tumor DNA to monitor metastatic breast
cancer. N. Engl. J. Med. 368,11991209 (2013).
39. M. J. Bamshad, J. A. Shendure, D. Valle, A. Hamosh, J. R. Lupski, R. A. Gibbs, E. Boerwinkle,
R. P. Lifton, M. Gerstein, M. Gunel, S. Mane, D. A. Nickerson; Centers for Mendelian Geno-
mics, The Centers for Mendelian Genomics: A new large-scale initiative to identify the
genes under lying rare Mendelian cond itions. Am.J.Med.Genet.A158A,15231525
(2012).
40. E. A. Worthey, A. N. Mayer, G. D. Syverson, D. Helbling, B. B. Bonacci, B. Decker, J. M. Serpe,
T. Dasu, M. R. Tschannen, R. L. Veith, M. J. Basehore, U. Broeckel, A. Tomita-Mitchell, M.J. Arca,
J. T. Casper, D. A. Margolis, D. P. Bick, M. J. Hessner, J. M. Routes, J. W. Verbsky, H. J. Jacob,
D. P. Dimmock, Making a definit ive diagnosis: Successful clinic al application of whole
exome sequencing in a child with intractable inflammatory bowel disease. Genet. Med.
13, 255262 (2011).
41. A.C.Need,V.Shashi,Y.Hitomi,K.Schoch,K.V.Shianna,M.T.McDonald,M.H.Meisler,
D. B . Goldstein, Clinical application of exome sequencing in undiagnosed genetic conditions.
J. Med. Genet. 49, 353361 (2012).
42. C. Gilissen, A. Hoischen, H. G. Brunner, J. A. Veltman, Disease gene identification strate-
gies for exome sequencing. Eur. J. Hum. Genet. 20, 490497 (2012).
43. Secretarys Advisory Committee on Heritable Disorders in Newborns and Children,
Newborn screening: Towards a uniform screening panel and system; http://www.h rsa.
gov/advisorycommittees/mchbadvisory/heritabledisorders/uniformscreening.pdf.
44. A. J. Goldenberg, R. R. Sharp, The ethical hazards and programmatic challenges of ge-
nomic newborn screening. JAMA 307, 461462 (2012).
45. Y. M. Lo, N. Corbetta, P. F. Chamberlain, V. Rai, I. L. Sargent, C. W. Redman, J. S. Wainscoat,
Presence of fetal DNA in maternal plasma and serum. Lancet 350, 485487 (1997).
46. H. C. Fan, Y. J. Blumenfeld, U. Chitkara, L. Hudgins, S. R. Quake, Noninvasive diagnosis of
fetal aneuploidy by shotgun sequencing DNA from maternal blood. Proc. Natl. Acad. Sci.
U.S.A. 105, 1626616271 (2008).
47. R. W. Chiu, K. C. Chan, Y. Gao, V. Y. Lau, W. Zh eng, T. Y. Leung, C. H. Foo, B. Xie, N. B. Tsui,
F. M. Lun, B. C. Zee , T. K. Lau, C. R. C antor, Y. M. Lo, N oninvasive prenatal diagnos is of
fetal chrom osomal aneu ploidy by massively p arallel gen omic sequencing of DNA i n
maternal plasm a. Proc. Natl. Ac ad. Sci. U.S.A. 105,20458204 63 (2008).
48. D. W. Bianchi, From prenatal genomic diagnosis to fetal personalized medicine: Progress
and challenges. Nat. Med. 18, 10411051 (2012).
49. Sequenom Inc., Sequenom, Inc. reports financial results for the third quarter of 2012 and
announces 90,000 annualized runrate for Maternit21 PLUS tests; http://sequenom.investorroom.
com/2012-11-08-Sequenom-Inc.-Reports-Financial-Results-For-The-Third-Quarter-Of-2012-
And-Announces-90-000-Annualized-Run-Rate-For-MaterniT21-PLUS-Tests.
50. Y. M. Lo, K. C. Chan, H. Sun, E. Z. Chen, P. Jiang, F. M. Lun, Y. W. Zheng, T. Y. Leung, T. K. Lau,
C. R. Cantor, R. W. Chiu, Maternal plasma DNA sequencing reveals the genome-wide ge-
netic and mutational profile of the fetus. Sci. Transl. Med. 2, 61ra91 (2010).
51. J. O. Kitzman, M . W. Snyder, M. Ven tura, A. P. Lewis , R. Qiu, L. E. Simm ons, H. S. Gammil l,
C. E. Rubens, D. A. Santillan, J. C. Murray, H. K. Tabor, M. J. Bamshad, E. E. Eichler, J. Shendure,
Noninvasive whole-genome sequencing of a human fetus. Sci. Transl. Med. 4, 137ra 76
(2012).
52. H. C. Fan, W. Gu, J. Wang, Y. J. Blumenfeld, Y. Y. El-Sayed, S. R. Quake, Non-invasive pre-
natal measurement of the fetal genome. Nature 487, 320324 (2012).
53. H. C. Fan, W. Gu, J. Wang, Y. J. Blume nfeld, Y. Y. El-Sayed, S. R. Quake, Errat um: Non-
invasive prenatal measurement of the fetal genome. Nature 489, 326 (2012).
54. McKusick-Nathans Institute of Genetic Medicine, Online Mendelian Inheritance in Man,
OMIM; http://omim.org/ [accessed 28 December 2012].
55. M. M. Kaback, Population-based genetic screening for reproductive counseling: The Tay-Sachs
disease model. Eur. J. Pediatr. 159 (Suppl. 3), S192S195 (2000).
56. C. J. Bell, D. L. Dinwiddie, N. A. Miller, S. L. Hateley, E. E. Ganusova, J. Mudge, R. J. Langley,
L. Zhang, C. C. Lee, F. D. Schi lkey, V. Sheth, J. E. Woodward, H. E. Peck ham, G. P. Schroth,
R. W. Kim, S. F. Kingsm ore, Carrier test ing for severe ch ildhood recessi ve diseases by
next-generat ion sequencing . Sci. Transl. Me d. 3, 65ra4 (2011).
57. C. S. Ku, E. K. Tan, D. N. Cooper, From the periphery to centre stage: De novo single nu-
cleotide variants play a key role in human genetic disease. J. Med. Genet. 50, 203211
(2013).
58. A. Kiezun, K. Garimella, R. Do, N . O. Stitziel, B. M . Neale, P. J. McLaren, N. Gupta, P. S klar,
P. F. Sullivan, J. L. Moran, C. M. Hultman, P. Lichtenstein, P. Magnusson, T. Lehner, Y. Y. Shugart,
A. L. Price, P. I. de Bakker, S. M. Purcell, S. R. Sunyaev, Exome sequencing and the genetic
basis of complex traits. Nat. Genet. 44, 623630 (2012).
59. M. J. Emond, T. Louie, J. Emerson, W. Zhao, R. A. Mathias, M. R. Knowles, F. A. Wright, M. J. Rieder,
H. K. Tabor, D. A. Nickerson, K. C. Barnes; National Heart, Lung, and Blood Institute (NHLBI)
GO Exome Sequencing Project; G. O. Lung, R. L. Gibson, M. J. Bamshad, Exome sequencing of
extreme phenotypes identifies DCTN4 as a modifier of chronic Pseu domona s aeru ginosa in-
fectionincysticfibrosis.Nat. Genet. 44,886889 (2012).
60. D. H. Roukos, E. Briasoulis, Indi vidualized preventive and therapeutic management of
hereditary breast ovarian cancer syndrome. Nat. Clin. Pract. Oncol. 4, 578590 (2007).
61. U. Ladabaum, G. Wang, J. Terdiman, A. Blanco, M. Kuppermann, C. R. Boland, J. Ford, E. Elkin,
K. A. Phillips, Strategies to identify the Lynch syndrome among patients with colorectal
cancer: A cost-effectiveness analysis. Ann. Intern. Med. 155,6979 (2011).
62. M. Chang, P. H. Green, Genetic testing before serologic screening in relatives of patients
with celiac disease as a cost containment method. J. Clin. Gastroenterol. 43,4350 (2009).
63. C. B. Do, D. A. Hinds, U. Francke, N. Eriksson, Comparison of family history and SNPs for
predicting risk of complex disease. PLoS Genet. 8, e1002973 (2012).
64. S . Mallal, E. Phillips, G. Carosi, J. M. Molina, C. Workman, J. Tomazic, E. Jägel-Guedes, S. Rugina,
O. Kozyrev, J. F. Cid, P. Hay, D. Nolan, S. Hughes, A. Hughes, S. Ryan, N. Fitch, D. Thorborn,
A. Benbow; PREDICT-1 Study Team, HLA-B*5701 screening for hypersensitivity to abacavir.
N. Engl. J. Med. 358,568579 (2008).
65. W. H. Chung, S. I. Hung, H. S. Hong, M. S. Hsih, L. C. Yang, H. C. Ho, J. Y. Wu, Y. T. Chen,
Medical genetics: A marker for StevensJohnson syndrome. Nature 428, 486 (2004).
66. C. J. Ross, H. Katzov-Eckert, M. P. Dubé, B. Brooks, S. R. Rassekh, A. Barhdadi, Y. Feroz-Zada,
H. Visscher, A. M. Brown, M. J. Rieder, P. C. Rogers, M. S. Phillips, B. C. Carleton, M. R. Ha yden;
CPNDS Consortium, Genetic variants in TPMT and COMT are associated with hearing loss in
children receiving cisplatin chemotherapy. Nat. Genet. 41,13451349 (2009).
67. S. Rost, A. Fregin, V. Ivaskevicius, E. Conzelmann, K. Hörtnagel, H. J. Pelz, K. Lappegard,
E. Seifried, I. Scharrer, E. G. Tuddenham, C. R. Müller, T. M. Strom, J. Oldenburg, Mutations in
VKORC1 cause warfarin resistance and multiple coagulation factor deficiency type 2. Nature
427,537541 (2004).
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 14
68. G. P. Aithal, C. P. Day, P. J. Kesteven, A. K. Daly, Assoc iation of polymorphisms in th e
cytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleeding compli-
cations. Lancet 353, 717719 (1999).
69. M. V. Holmes, P. Perel, T. Shah, A. D. Hingorani, J. P. Casas, CYP2C19 genotype, clopidogrel
metabolism, platelet function, and cardiovascular events: A systematic review and meta-
analysis. JAMA 306, 27042714 (2011).
70. D. R. Booth, G. Ahlenstiel, J. George, Pharmacogenomics of hepatitis C infections: Person-
alizing therapy. Genome Med. 4, 99 (2012).
71. Z. Zhang, An in vitro diagnostic multivariate index assay (IVDMIA) for ovarian cancer:
Harvesting the power of multiple biomarkers. Rev. Obstet. Gynecol. 5,3541 (2012).
72. M. Dowsett, J. Cuzick, C. Wale, J. Forbes, E. A. Mallon, J. Salter, E. Quinn, A. Dunbier, M. Baum,
A. Buzdar, A. Howell, R. Bugarini, F. L. Baehner, S. Shak, Prediction of risk of distant recurrence
using the 21-gene recurrence score in node-negative and node-positive postmenopausal
patients with breast cancer treated with anastrozole or tamoxifen: A TransATAC study. J. Clin.
Oncol. 28, 18291834 (2010).
73. K.S. Albain,W.E.Barlow,S.Shak,G.N.Hortobagyi,R.B.Livingston,I.T.Yeh,P.Ravdin,
R.Bugarini,F.L.Baehner,N.E.Davidson,G.W.Sledge,E.P.Winer,C.Hudis,J.N.Ingle,
E. A. Perez, K. I. Pritchard, L. Shepherd, J. R. Gralow, C. Yoshizawa, D. C. Allred, C. K. Osborne,
D. F. Hayes; Breast Cancer Intergroup of North America , Prognostic and predictive value of
the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-
receptor-positive breast cancer on chemotherapy: A retrospective analysis of a randomised
trial. Lancet Oncol. 11,5565 (2010).
74. S. Paik, G. Tang, S. Shak, C.Kim, J. Baker, W. Kim, M. Cronin, F. L. Baehner, D.Watson, J. Bryant,
J. P. Costantino, C. E. Geyer Jr., D. L. Wickerham, N. Wolmark, Gene expression and benefit of
chemotherapy in women with node-negative, estrogen receptorpositive breast cancer.
J. Clin. Oncol. 24, 37263734 (2006).
75. S. Paik, S. Shak, G. Tang, C. Kim, J. Baker, M. Cronin, F. L. Baehner, M. G. Walker, D. Watson,
T. Park, W. Hiller, E. R. Fisher, D. L. Wickerham, J. Bryant, N. Wolmark, A multigene assay to
predict recurrence of tamoxife n-treated, node-negative bre ast cancer. N. Engl. J. Med.
351, 28172826 (2004).
76. U.S. Food and Drug Administration, Draft guidance for industry, clinical laboratories, and
FDA staff: In vitro diagnostic multivariate index assays; http://www.fda.gov/downloads/
MedicalDevices/.../ucm071455.pdf.
77. H.A.AzimJr.,S.Michiels,F.Zagouri,S.Delaloge,M.Filipits,M.Namer,P.Neven,W.F.Symmans,
A.Thompson,F.André,S.Loi,C.Swanton,Utilityofprognosticgenomictestsinbreastcancer
practice: The IMPAKT 2012 Working Group Consensus Statement. Ann. Oncol. 24,647654
(2013).
78. S. Rosenberg, M. R. Elashoff, P. Beineke, S. E. Daniels, J. A. Wingrove, W. G. Tingley, P. T. Sager,
A. J. Sehnert, M. Yau, W. E. Kraus, L. K. Newby, R. S. Schwartz, S. Voros, S. G. Ellis, N. Tahirkheli,
R. Waksman, J. McPherson, A. Lansky, M. E. Winn, N. J. Schork, E. J. Topol; PREDICT (Person-
alized Risk Evaluation and Diagnosis in the Coronary Tree) Investigators, Multicenter valida-
tion of the diagnosti c accuracy of a blood-ba sed gene expression te st for assessing
obstructive coronary artery disease in nondiabetic patients. Ann. Intern. Med. 153, 425434
(2010).
79. U.S. Centers for Medicare and Medicaid Services, New and reconsidered clinical labora-
tory fee schedule; http://www.cms.gov/Medicare/Medicare-Fee-for-Service-Payment/
ClinicalLabFeeSched/Downloads/CLFS-CY2013-Preliminary-Payment-Determinations.pdf.
80. G. M. Weinstock, Genomic approaches to studying the human microbiota. Nature 489,
250256 (2012).
81. D. A. Relman, Microbial genomics and infectious diseases. N. Engl. J. Med. 365, 347357
(2011).
82. Human Microbiome Project Consortium, Structure, function and diversity of the healthy
human microbiome. Nature 486, 207214 (2012).
83. J. Qin, Y. Li, Z. C ai, S. Li, J. Zhu, F. Zhang, S. Liang, W. Zhang, Y. Guan, D. Shen, Y. Peng,
D. Zhang, Z. Jie, W. Wu, Y. Qin, W. Xue, J. Li, L. Han, D. Lu, P. Wu, Y. Dai, X. Sun, Z. Li, A. Tang,
S. Zhong, X. Li, W. Chen, R. Xu, M. Wang, Q. Feng, M. Gong, J. Yu, Y. Zhang, M. Zhang,
T. Hansen, G. Sanchez, J. Raes, G. Falony, S. Okuda, M. Almeida, E. LeChatelier, P. Renault,
N. Pons, J. M. Batto, Z. Zhang, H. Chen, R. Yang, W. Zheng, S. Li, H. Yang, J. Wang, S. D. Ehrlich,
R. Nielsen, O. Pedersen, K. Kristiansen, J. Wang, A metagenome-wide association study of gut
microbiota in type 2 diabetes. Nature 490,5560 (2012).
84. I. Cho, M. J. Blaser, The human microbiome: At the interface of health and disease. Nat.
Rev. Genet. 13, 260270 (2012).
85. K. J. Maloy, F. Powrie, Intestinal homeostasis and its breakdown in inflammatory bowel
disease. Nature 474, 298306 (2011).
86. I. Mukhopadhya, R. Hansen, E. M. El-Omar, G. L. Hold, IBDWhat role do Proteobacteria
play? Nat. Rev. Gastroenterol. Hepatol. 9, 219230 (2012).
87. T. J. Borody, A. Khoruts, Fecal microbiota transplantation and emerging applications. Nat.
Rev. Gastroenterol. Hepatol. 9,8896 (2011).
88. M. A. Marra, S. J. Jones, C. R. Astell, R. A. Holt, A. Brooks-Wilson, Y. S. Butterfield, J. Khattra,
J. K. Asano, S. A. Barber, S. Y. Chan, A. Cloutier, S. M. Coughlin, D. Freeman, N. Girn, O. L. Griffith,
S. R. Leach, M. Mayo, H. McDonald, S. B. Montgomery, P. K. Pandoh, A. S. Petrescu, A. G. Robertson,
J. E. Schein, A. Siddiqui, D. E. Smailus, J. M. Stott, G. S. Yang, F. Plummer, A. Andonov,
H. Art sob, N. Bastien, K. Bernard, T. F. Booth, D. Bowness, M. Czub, M. Drebot, L. Fernando,
R. Flick, M. Garbutt, M . Gray, A. Grolla , S. Jones, H. Feld mann, A. Meyers , A. Kabani, Y. Li,
S. Normand, U. Stroher, G. A. Tipples, S. Tyler, R. Vogrig, D. Ward, B. Watson, R. C. Brunham,
M. Krajden, M. Petric, D. M. Skowronski, C. Upton, R. L. Roper, The genome sequence of the
SARS-associated coronavirus. Science 300,13991404 (2003).
89. C. S. Chin, J. Sorenson, J. B. Harris, W. P. Robins, R. C. Charles, R. R. Jean-Charles, J. Bullard,
D. R.Webster,A.Kasarskis,P.Peluso,E.E.Paxinos,Y.Yamaichi,S.B.Calderwood,J.J.Mekalanos,
E. E. Schadt, M. K. Waldor, The origin of the Haitian cholera outbreak strain. N. Engl. J. Med. 364,
3342 (2011).
90. J. L. Gardy, J. C. Johnston, S. J. Ho Sui, V. J. Cook, L. Shah, E. Brodkin, S. Rempel, R. Moore,
Y. Zhao, R. Holt, R. Varhol, I. Birol, M. Lem, M. K. Sharma,K. Elwood, S. J. Jones, F. S. Brinkman,
R. C. Brunham, P. Tang, Whole-genome sequencing and social-network analysis of a tuber-
culosis outbreak. N. Engl. J. Med. 364,730739 (2011).
91. G . J. Smith, D. Vijaykrishna, J. Bahl, S. J. Lycett, M. Worobey, O. G. Pybus, S. K. Ma, C. L. Cheung,
J. Raghwani, S. Bhatt, J. S. Peiris, Y. Guan, A. Rambaut, Origins and evolutionary genomics of
the 2009 swine-origin H1N1 influenza A epidemic. Nature 459, 11221125 (2009).
92. E. S. Snitkin, A. M. Zelazny, P. J. Thomas, F. Stock; NISC Comparative Sequencing Program
Group,D.K.Henderson,T.N.Palmore,J.A.Segre,Trackingahospitaloutbreakofcarbapenem-
resistant Klebsiella pneumoniae with whole-genome sequencing. Sci. Transl. Med. 4, 148ra116
(2012).
93. E. D. Green, M. S. Guyer; National Human Genome Research Institute, Charting a course
for genomic medicine from base pairs to bedside. Nature 470, 204213 (2011).
94. J. E. Haddow, G. E. Palomaki, in Human Genome Epidemiology: A Scientific Foundation for
Using Genetic Information to Improve Health and Prevent Disease,M.Khoury,J.Little,
W. Burke, Eds. (Oxford Univ. Press, New York, 2003), pp. 217233.
95. S. R. Tunis, D. B. Stryer, C. M. Cl ancy, Practical clinical tria ls: Increasin g the value of clin-
ical research for decision makin g in clinical an d health policy. JAMA 290, 162 41632
(2003).
96. D. A. Mrazek, C. Lerman, Facilitating clinical implementation of pharmacogenomics. JAMA
306, 304305 (2011).
97. G. S. Gins burg, J. Staples, A. P. Aberne thy, Academic medi cal centers: Ripe fo r rapid-
learning personalized health care. Sci. Transl. Med. 3, 101cm27 (2011).
98. G. S. Ginsbu rg, N. M. Kuderer, Co mparative effectiven ess research, genom ics-enabled
personalized medicine, and rapid learning health care: A common bond. J. Clin. Oncol.
30, 42334242 (2012).
99. M.W.Foster,J.J.Mulvihill,R.R.Sharp,Evaluatingtheutilityofpersonalgenomic
information. Genet. Med. 11, 570574 (2009).
100.S.D.Grosse,C.M.McBride,J.P.Evans,M. J.Khoury,Personalutilityandgenomic
information: Look before you leap. Genet. Med. 11, 575576 (2009).
101. J. R. Botkin, S.M. Teutsch, C. I. Kaye, M. Hayes, J. E.Haddow, L. A. Bradley, K.Szegda, W. D. Dotson;
EGAPP Working Group, Outcomes of interest in evidence-based evaluations of genetic tests.
Genet. Med. 12,228235 (2010).
102. M. J. Khoury, C. M. McBride, S. D. Schully, J. P. Ioannidis, W. G. Feero, A. C. Janssens, M. Gwinn,
D. G. Simons-Morton, J. M. Bernhardt, M. Cargill, S. J. Chanock, G. M. Church, R. J. Coates,
F. S. Collins, R. T. Croyle, B. R. Davis, G. J. Downing, A. Duross, S. Friedman, M. H. Gail,
G. S. Ginsburg, R. C. Green, M. H. Greene, P. Greenland, J. R. Gulcher, A. Hsu, K. L. Hudson,
S.L.Kardia,P.L.Kimmel,M.S.Lauer,A.M.Miller,K.Offit,D.F.Ransohoff,J.S.Roberts,
R. S. Rasooly, K. Stefansson, S. F. Terry, S. M. Teutsch, A. Trepanier, K. L. Wanke, J. S. Witte,
J. Xu; Centers for Disease Control and Prevention, The Scientific Foundation for personal
genomics: Recommendations from a Natio nal Institutes of HealthCenters for Disease
Control and Prevention multidisciplinary workshop. Genet. Med. 11, 559567 (2009).
103. U.S. Food and Drug Administration, Table of pharmacogenomic biomarkers in drug
labels; http://www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/
ucm083378.htm.
104. C. F. Thorn, T. E. Klein, R. B. Altman, Pharmacogenomics and bioinformatics: PharmGKB.
Pharmacogenomics 11, 501505 (2010).
105. W. S. Rubinstein, D. R. Maglott, J. M. Lee, B. L. Kattman, A. J. Malheiro, M. Ovetsky, V. Hem,
V. Gorelenkov, G. Song, C. Wallin, N. Husain, S. Chitipiralla, K. S. Katz, D. Hoffman, W. Jang,
M. Johnson, F. Karmanov, A. Ukrainchik, M. Denisenko, C. Fomous, K. Hudson, J. M. Ostell,
The NIH genetic testing registry: A new, centralized database of genetic tests to enable
access to comprehensive information and improve transparency. Nucleic Acids Res. 41,
D925D935 (2013).
106. U.S. Centers for Medicare and Medicaid Services, Update on horizon scans of genetic tests
currently available for clinical use in cancers; http://www.cms.go v/Medicare/Coverage/
DeterminationProcess/downloads/id81TA.pdf.
107. U.S. Centers for Medicare and Medicaid Services, Update on mapping the landscape of
genetic tests for non-cancer diseases/conditions; http://www.cms.gov/Medicare/Coverage/
DeterminationProcess/downloads/id86TA.pdf.
108. A. O. Berg, The CDCs EGAPP initiative: Evaluating the clinical evidence for genetic tests.
Am. Fam. Physician 80, 1218 (2009).
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 15
109. M. V. Relling, T. E. Klein, CPIC: Clinical Pharmacogenetics Implementation Consortium of
the Pharmacogenomics Research Network. Clin. Pharmacol. Ther. 89, 464467 (2011).
110. M. S. Williams, in Genomic and Personalized Medicine,G.Ginsburg,H.Willard,Eds.(Elsevier,
New York, ed. 2, 2013), vol. 1, pp. 340352.
111. J. E. Dancey, P. L. Bedard, N. Onetto, T. J. Hudson, The genetic basis for cancer treatment
decisions. Cell 148, 409420 (2012).
112. L. Goetz, K. Bethel, E. J. Topol, Rebooting cancer tissue handling in the sequencing era:
Toward routine use of frozen tumor tissue. JAMA 309,3738 (2013).
113. G. Gerhard, D. J . Carey, G. D. Stee le, in Genomic and Personalized Medicine, G. Ginsbur g,
H. Willard, Eds . (Elsevier, New Y ork, ed. 2, 2013), vol. 1, pp. 287294 .
114. K. Ka wamoto, D. F. L obach, H. F. Will ard, G. S. Gin sburg, A national clinical decis ion
support infras tructure to ena ble the widespre ad and consistent practice of genomic
and personalized medicine. BMC Med. Inform. Decis. Mak. 9, 17 (2009).
115. J. Starren, M. S. Williams, E. P. Bottinger, Crossing the omic chasm: A time for omic an-
cillary systems. JAMA 309, 12371238 (2013).
116. Health Level 7, Im plementation guide for CDA rel ease 2 genetic testing report (G TR);
http://www.hl7.org/documentcenter/public_temp_965C9715-1C23-BA17-0C02E787A-
BE1A9D8/wg/clingenomics/docs/Genetic Testing Report (GTR) - 2012.10.30.pdf.
117. J. Woodcock, in Genomic and Personalized Medicine, G. Ginsburg, H. Willard, Eds. (Elsevier,
New York, ed. 2, 2013), pp. 422432.
118. U.S. Food and Drug Administration, Class II special controls guidance document: Drug
metabolizing enzyme genotyping systemGuidance for industry and FDA staff; http://
www.fda.gov/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/
ucm077933.htm.
119. U.S. Food and Drug Administration, Guidance for industry and Food and Drug Administra-
tion staffFactorstoconsiderwhenmakingbenefit-riskdeterminationsinmedicaldevice
premarket approvals and de no vo classifications; http://www .fda.gov/MedicalDevices/
DeviceRegulationandGuidance/GuidanceDocuments/ucm267829.htm.
120. U.S. National Institutes of Health, Direct-to-consumer genetic testing: Report of the Sec-
retarys Advisory Committee on Genetics, Health, and Society; http://oba.od.nih.gov/oba/
sacghs/reports/SACGHS_DTC_Report_2010.pdf.
121. U.S. National Institutes of Health, U.S. system of oversight of genetic testing: A response
to the charge of the Secretary of Health and Human Services; http://oba.od.nih.gov/oba/
SACGHS/reports/SACGHS_oversight_report.pdf.
122. U.S. Food and Drug Administration, Oversight of laboratory developed tests; Public
meeting; Req uest for commen ts; http:// www.gpo.gov /fdsys/pkg /FR-2010-0 6-17/html/
2010-14654.htm.
123. A. Hresko, S. B. Haga, Insurance coverage policies for personalized medicine. J. Pers. Med.
2, 201216 (2012).
124. U.S. Centers for Medicare and Medicaid Services, MEDCAC Meeting 2/25/2009Genetic
(genomic) testing; http://www.cms.gov/medicare-coverage-database/details/medcac-
meeting-details.aspx?MEDCACId=47&NCAId=224&ver=15&NcaName=Pharmacogenomic+
Testing+for+Warfarin+Response&bc=ACAAAAAAEAAA&.
125. New and reconsidered clinical laboratory fee schedule test codes and preliminary payment
determinations; http://www.cms.gov/Center/Provider-Type/Clinical-Labs-Center.html.
126. R. Cook-Deegan, Patent law. Law and science collide over human gene patents. Science
338, 745747 (2012).
127. M. Gymrek, A. L. McGuire, D. Golan, E. Halperin, Y. Erlich, Identifying personal genomes by
surname inference. Science 339, 321324 (2013).
128. J. Kaye, P . Boddin gton, J. d e Vries, N . Hawkin s, K. Melh am, Ethi cal impl icatio ns of the
use of whole genome methods in medical research. Eur. J. Hum. Genet. 18, 398403
(2010).
129. J. S. King, Genetic tests: Politics and fetal diagnostics collide. Nature 491,3334 (2012).
130. S. B. Haga, in Genomic and Personalized Medicine, G. Ginsburg, H. Willard, Eds. (Elsevier,
New York, ed. 2, 2013), vol. 1, pp. 392404.
131. American College of Medical Genetics, Points to consider in the clinical application of
genomic sequencing; http://www.acmg.net/StaticContent/PPG/Clinical_Appli cation_of_
Genomic_Sequencing.pdf.
132. American College of Medical Genetics, ACMG recommendations for reporting of incidental
findings in clinical exome and genome sequencing; http://www.acmg.net/docs/ACMG_
Releases_Highly-Anticipated_Recommendations_on_Inci dental_Findings_in_Clinical_
Exome_and_Genome_Sequencing.pdf.
133. American College of Medical Genetics, Incidental findings in clinical genomics: A clarification;
http://www.acmg.net/docs/Incidental_Findings_in_Clinical_Genomics_A_Clarification.pdf.
134. U.S. National Human Genome Research Institute, Characterizing and displaying genetic
variants for clinical action workshop; http://www.genome.gov/27546546.
135. R. A. Schnoll, A. E. Shields, Physician barriers to incorporating pharmacogenetic treatment
strategies for nicotine dependence into clinical p ractice. Clin. Pharmacol. Ther. 89, 345347
(2011).
136. S. B. Haga, W. Burke, G. S. Ginsburg, R. Mills, R. Agans, Primary care physiciansknowledge
of and experience with pharmacogenetic testing. Clin. Genet. 82, 388394 (2012).
137. K. P. Powell, W. A. Cogswell, C. A. Christianson, G. Dave, A. Verma, S. Eubanks, V. C. Henrich,
Primary care physiciansawareness, experience and opinions of direct-to-consumer genetic
testing. J. Genet. Couns. 21, 113126 (2012).
138. E. J. Houwink, S. J. van Luijk, L. Henneman, C. van der Vleuten, G. Jan Dinant, M. C. Cornel,
Genetic educational needs and the role of genetics in primary care: A focus group study
with multiple perspectives. BMC Fam. Pract. 12, 5 (2011).
139. U.S. National Institutes of Health, Genetics education and training: Report of the Secre-
tarys Advisory Committee on Genetics, Health, and Society; http://oba.od.nih.gov/oba/
SACGHS/reports/SACGHS_education_report_2011.pdf.
140. National Coalition for Health Professionals Education in Genetics, Core competencies for
all health professionals; http://www.nchpeg.org/index.php?option=com_content&view=
article&id=237&Itemid=84.
141. J. Magee, J. I. Gordon, A. Whelan, Bringi ng the human genome and the revolution in
bioinformatics to the medical school classroom: A case report from Washington Univer-
sity School of Medicine. Acad. Med. 76, 852855 (2001).
142. B. R. Korf, Integration of genetics into clinical teaching in medical school education. Genet.
Med. 4, 33S38S (2002).
143. C. M. Wiener, P. A. Thomas, E. Goodspeed, D. Valle, D. G. Nichols, Genes to society”—The
logic and process of the new curriculum for the Johns Hopkins University School of Med-
icine. Acad. Med. 85, 498506 (2010).
144. S. U. Dhar, R. L. Alford, E. A. Nelson, L. Potocki, Enhancing exposure to genetics and ge-
nomics through an innovative medica l school curriculum. Genet. Med. 14,163167 (2012).
145. The Health Literacy of Americas Adults: Results from the 2003 National Assessment of Adult
Literacy (National Center for Education Statistics, Washington, DC, 2006).
146. D. H. Lea, K. A. Kaphingst, D. Bowen, I. Lipkus, D. W. Hadley, Communicating genetic and
genomic information: Health literacy and numeracy considerations. Public Health Genomics
14,279289 (2011).
147. G. S. Ginsburg, J. J. McCarthy, Personalized medicine: Revolutionizing drug discovery and
patient care. Trends Biotechnol. 19, 491496 (2001).
148. D. Wu, A. Sherwood, J. R. Fromm, S. S. Winter, K. P. Dunsmore, M. L. Loh, H. A. Greisman,
D. E. Sabath, B. L. Wood, H. Robins, High-throughput sequencing detects minimal residual
disease in acute T lymphoblastic leukemia. Sci. Transl. Med. 4, 134ra63 (2012).
149. A. C. Loga n, H. Gao, C . Wang, B. S ahaf, C. D . Jones, E. L. Marsh all, I. Bu ño, R. Armstrong ,
A.Z.Fire,K.I.Weinberg,M.Mindrinos,J.L.Zehnder,S.D.Boyd,W.Xiao,R.W.Davis,
D. B . Miklos, High-throughput VDJ sequencing for quantification of minimal residual disease
in chronic lymphocytic leukemia and immune reconstitution assessment. Proc. Natl. Acad. Sci.
U.S.A. 108,2119421199 (2011).
150. N. Jiang, J. He, J. A. We instei n, L. Penl and, S. Sa saki, X. S . He, C. L. Dek ker, N. Y. Z heng,
M. Huang, M. Sullivan, P. C. Wilson, H. B. Greenberg, M. M. Davis, D. S. Fisher, S. R. Quake,
Lineage structure of the human antibody repertoire in response to influenza vaccination.
Sci. Transl. Med. 5, 171ra19 (2013).
151. T. M. Snyder, K. K. Khush, H. A. Valantine, S. R. Quake, Universal noninvasive detection of
solid organ transplant rejection. Proc. Natl. Acad. Sci. U.S.A. 108, 62296234 (2011).
152. E. E. Schadt, S. Turner, A. Kasarskis, A window into third-generation sequencing. Hum.
Mol. Genet. 19, R227R240 (2010).
153. International Cancer Genome Consortium; T. J. Hudson, W. Anderson, A. Artez, A. D. Barker,
C. Bell, R. R. Bernabé, M. K. Bhan, F. Calvo, I. Eerola, D. S. Gerhard, A. Guttmacher, M. Guyer,
F. M. Hemsley, J. L. Jennings, D. Kerr, P. Klatt, P. Kolar, J. Kusada, D. P. Lane, F. Laplace,
L. Youyong, G. Nettekoven, B. Ozenberger, J. Peterson, T. S. Rao, J. Remacle, A. J. Schafer,
T. Shibata, M. R. Stratton, J. G. Vockley, K. Watanabe, H. Yang, M. M. Yuen, B. M. Knoppers,
M. Bobrow, A. Cambon-Thomsen, L. G. Dressler, S. O. Dyke, Y. Joly, K. Kato, K. L. Kennedy,
P. Nicolás, M. J. Parker, E. Rial-Sebbag, C. M. Romeo-Casabona, K. M. Shaw, S. Wallace,
G. L. Wiesner,N.Zeps,P.Lichter,A.V.Biankin,C.Chabannon,L.Chin,B.Clément,E.deAlava,
F.Degos,M.L.Ferguson,P.Geary,D.N.Hayes,T.J.Hudson,A.L.Johns,A.Kasprzyk,
H. Nakagawa, R. Penny, M. A. Piris, R. Sarin, A. Scarpa, T. Shibata, M. van de Vijver, P. A. Futreal,
H. Aburatani, M. Bayés, D. D. Botwell, P. J. Campbell, X. Estivill, D. S. Gerhard, S. M. Grimmond,
I. Gut, M. Hirst, C. pez-Otín, P. Majumder, M. Marra, J. D. McPherson, H. Nakagawa, Z. Ning,
X.S.Puente,Y.Ruan,T.Shibata,M.R.Stratton,H.G.Stunnenberg,H.Swerdlow,V.E.Velculescu,
R. K. Wilson, H. H. Xue, L. Yang, P. T. Spellman, G. D. Bader, P. C. Boutros, P. J. Campbell, P. Flicek,
G. Getz, R. Guigó, G. Guo , D. Haussler, S. Heath, T. J. Hubbar d, T. Jiang, S. M. Jon es, Q. Li,
N. López-Bigas, R. Luo, L. Muthuswamy, B. F. Ouellette, J. V. Pearson, X. S. Puente, V. Quesada,
B.J.Raphael,C.Sander,T.Shibata,T.P.Speed,L.D.Stein,J.M.Stuart,J.W.Teague,Y.Totoki,
T.Tsunoda,A.Valencia,D.A.Wheeler,H.Wu,S.Zhao,G.Zhou,L.D.Stein,R.Guigó,T.J.Hubbard,
Y. Joly, S. M. Jones, A. Kasprzyk, M. Lathrop, N. López-Bigas, B. F. Ouellette, P. T. Spellman,
J. W.Teague,G.Thomas,A.Valencia,T.Yoshida,K.L.Kennedy,M.Axton,S.O.Dyke,P.A.Futreal,
D. S. Gerhard, C. Gunter, M. Guyer, T. J. Hudson, J. D. McPherson, L. J. Miller, B. Ozenberger,
K. M. Shaw, A. Kasprzyk, L. D. Stein, J. Zhang, S. A. Haider, J. Wang, C. K. Yung, A. Cros, Y. Liang,
S. Gnaneshan, J. Guberman, J. Hsu, M. Bobrow, D. R. Chalmers, K. W. Hasel, Y. Joly, T. S. Kaan,
K. L. Kennedy, B. M. Knoppers, W. W. Lowrance, T. Masui, P. Nicolás, E. Rial-Sebbag,
L. L . Rodriguez, C. Vergely, T. Yoshida, S. M. Grimmond, A. V. Biankin, D. D. Bowtell, N. Cloonan,
A.deFazio,J.R.Eshleman,D.Etemadmoghadam,B.B.Gardiner,J.G.Kench,A.Scarpa,
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 16
R. L. Sutherland, M. A. Tempero, N. J. Waddell, P. J. Wilson, J. D. McPherson, S. Gallinger,
M. S. Tsao, P. A. Shaw, G. M. Petersen, D. Mukhopadhyay, L. Chin, R. A. DePinho, S. Thayer,
L. Muthuswamy, K. Shazand, T. Beck,M. Sam, L. Timms, V. Ballin, Y. Lu, J.Ji, X. Zhang, F. Chen,
X. Hu, G. Zhou, Q. Yang, G. Tian, L. Zhang, X. Xing, X. Li, Z. Zhu, Y. Yu, J. Yu, H. Yang,
M. Lathrop, J. Tost, P. Brennan, I. Holcatova, D. Zaridze, A. Brazma, L. Egevard, E. Prokhortchouk,
R. E. Banks, M. Uhlén, A. Cambon-Thomsen, J. Viksna, F. Ponten, K. Skryabin, M. R. Stratton,
P. A.Fu treal, E. Birney, A. Borg, A. L. Bør resen-Dale, C. Caldas, J. A. Foekens, S. Martin,
J. S. Reis-Filho, A. L. Richardson, C. Sotiriou, H. G. Stunnenberg, G. Thoms, M. van de Vijver,
L. vant Veer, F. Calvo, D. B irnb aum, H . Blan che, P. B ouch er, S. B oyault, C . Chab anno n,
I. Gut , J. D . Mass on-J acq uemi er, M. Lathrop, I. Pauporté, X. P ivot, A. Vincent -Salomon,
E. Tabone, C. Thei llet, G. Thomas, J. Tost, I. Treilleux, F. Calvo, P. Bioulac-Sage, B. Clément,
T. Decaens, F. Degos, D. Franco, I. Gut, M. Gut, S. Heath, M. Lathrop, D. Samuel, G. Thomas,
J.Zucman-Rossi,P.Lichter,R.Eils,B.Brors,J.O.Korbel,A.Korshunov,P.Landgraf,H.Lehrach,
S. Pfister, B. Radlwimmer, G. Reifenberger, M. D. Taylor, C. von Kalle, P. P. Majumder, R. Sarin,
T.S.Rao,M.K.Bhan,A.Scarpa,P.Pederzoli,R.A.Lawlor,M.Delledonne,A.Bardelli,A.V.Biankin,
S. M. Grimmond, T. Gress, D. Klimstra, G. Zamboni, T. Shibata, Y. Nakamura, H. Nakagawa,
J. Kusada, T. Tsunoda, S. Miyano, H. Aburatani, K. Kato, A. Fujimoto, T. Yoshida, E. Campo,
C. López-Otín, X. Estivill, R. Guigó, S. de Sanjoséé, M. A. Piris, E. Montserrat, M. González-Díaz,
X. S. Puente, P. Jares, A. Valencia, H. Himmelbauer, V. Quesada, S. Bea, M. R. Stratton,
P. A. Futreal, P. J. Campbell, A. Vincent-Salomon, A. L. Richardson, J. S. Reis-Filho, M. van de
Vijver, G. Thomas, J. D. Masson-Jacquemier, S. Aparicio, A. Borg, A. L. Børresen-Dale, C. Caldas,
J. A. Foekens, H. G. Stunnenberg, L. vant Veer, D. F. Easton, P. T. Spellman, S. Martin, A. D. Barker,
L.Chin,F.S.Collins,C.C.Compton,M.L.Ferguson,D.S.Gerhard,G.Getz,C.Gunter,
A. Guttmach er, M. Guyer, D. N. Ha yes, E. S. Lander , B. Ozenberger, R. Penny, J . Peterson,
C. Sander, K. M. Shaw, T. P. Speed, P. T. Spellman, J. G. Vockley, D. A. Wheeler, R. K. Wilson,
T.J.Hudson,L.Chin,B.M.Knoppers,E.S.Lander,P.Lichter,L.D.Stein,M.R.Stratton,
W. Anderson, A. D. Barker, C. Bell, M. Bobrow, W. Burke, F. S. Collins, C. C. Compton,
R. A. DePinho, D. F. Easton, P. A. Futreal, D. S. Gerhard, A. R. Green, M. Guyer, S. R. Hamilton,
T.J.Hubbard,O.P.Kallioniemi,K.L.Kennedy,T.J.Ley,E.T.Liu,Y.Lu,P.Majumder,
M.Marra,B.Ozenberger,J.Peterson,A.J.Schafer,P.T.Spellman,H.G.Stunnenberg,
B. J. Wainwright, R. K. Wilson, H. Yang, International network of cancer genome projects.
Nature 464,993998 (2010).
154. N. Erik sson , J. M. M acpherso n, J. Y. T ung, L . S. Hon, B. Na ught on, S. S axon ov, L. Av ey,
A. Wojcicki, I. Peer, J. Mountain, Web-based, participant-driven studies yield novel genetic
associations for common traits. PLoS Genet. 6, e1000993 (2010).
155. A. C. Janssens, P. Kraft, Research conducted using data obtained through online commu-
nities: Ethical implications of methodological limitations. PLoS Med. 9, e1001328 (2012).
156. R. R. Kalf, R. Bakker, A. C. Janss ens, Predictive ability of direct-to-consumer pharmaco-
genetic testing: Whe n is lack of evidence re ally lack of evidence ? Pharmacogenomics
14, 341344 (2013).
157. World Health Organization, International statistical classification of diseases and related
health problems 10th revision; http://apps.who.int/classifications/icd10/browse/2010/en.
158. E. Campo, S. H. Swerdlow, N. L. Harris, S. Pileri, H. Stein, E. S. Jaffe, The 2008 WHO clas-
sification of lymphoid neoplasms and beyond: Evolving concepts and practical applica-
tions. Blood 117, 50195032 (2011).
159. J. W. Vardiman, J. Thiele, D. A. Arber, R. D. Brunning, M. J. Borowitz, A. Porwit, N. L. Harris,
M. M. Le Beau, E. Hellström-Lindberg, A. Tefferi, C. D. Bloomfield, The 2008 revision of the
World Health Organization (WHO) classification of myeloid neoplasms and acute leuke-
mia: Rationale and important changes. Blood 114, 937951 (2009).
160. W. D. Travis, E. Brambilla, M. Noguchi, A. G. Nicholson, K. R. Geisinger, Y. Yatabe, D. G. Beer,
C.A.Powell,G.J.Riely,P.E.VanSchil,K.Garg,J.H.Austin,H.Asamura,V.W.Rusch,F.R.Hirsch,
G.Scagliotti,T.Mitsudomi,R.M.Huber,Y.Ishikawa,J.Jett,M.Sanchez-Cespedes, J.P.Sculier,
T. Takahashi, M. Tsuboi, J. Vansteenkiste, I. Wistuba, P. C. Yang, D. Aberle, C. Brambi lla,
D. Flieder, W. Franklin, A. Gazdar, M. Gould, P. Hasleton, D. Henderson, B. Johnson, D. Johnson,
K. Kerr, K. Kuriyama, J. S. Lee, V. A. Miller, I. Petersen, V. Roggli, R. Rosell, N. Saijo, E. Thunnissen,
M. Tsao, D. Yankelewitz, International Association for the Study of Lung Cancer/American
Thoracic Society/European Respiratory Society international multidisciplinary classification
of lung adenocarcinoma. J. Thorac. Oncol. 6,244285 (2011).
161. Toward Precision Medicine: Building a Knowledge Network for Biomedical Research and a
New Taxonomy of Disease (National Academies Press, Washington, DC, 2011).
162. World Economic Forum, Global agenda council on personalized & precision me dicine
2012-2013; http://www.weforum.org/content/global-agenda-council-personalized-
precision-medicine-2012.
163. A. N. Kho, J. A. Pacheco, P. L. Pei ssig, L. Rasmuss en, K. M. Newton, N. Weston, P. K. Crane,
J.Pathak,C.G.Chute,S.J.Bielinski,I.J.Kullo,R.Li,T.A.Manolio,R.L.Chisholm,J.C.Denny,
Electronic medical records for genetic research: Results of the eMERGE consortium. Sci.Transl.
Med. 3, 79re1 (2011).
164. B. Carlson, Vanderbilt pioneers bedside genetics. Biotechnol. Healthc. 9,3132 (2012).
165. E. Dolgin, Preemptive genotyping trialed to prevent adverse drug reactions. Nat. Med. 17,
1323 (2011).
166. V. Gubala, L. F. Harris, A. J. Ricco, M. X. Tan, D. E. Williams, Point of care diagnostics: Status
and future. Anal. Chem. 84, 487515 (2012).
167. E. J. Topol, The Creative Destruct ion of Medicine: How the Digital Revolution Will Create
Better Health Care (Basic Books, New York, 2012).
168. R. Chen, G. I. Mias, J. Li-Pook-Than, L. Jiang, H. Y. Lam, R. Chen, E. Miriami, K. J. Karczewski,
M. Hariharan, F. E. Dew ey, Y. Cheng, M. J. Clar k, H. Im, L. Habegger, S. Balasubramanian,
M. OHuallachain,J.T.Dudley,S.Hillenmeyer,R.Haraksingh,D.Sharon,G.Euskirchen,
P. Lacroute, K. Bettinger, A. P. Boyle, M. Kasowski, F. Grubert, S. Seki, M. Garcia, M. Whirl-Carrillo,
M. Gallardo, M. A. Blasco, P. L. Greenberg, P. Snyder, T. E. Klein, R. B. Altman, A. J. Butte, E. A. Ashley,
M. Gerstein, K. C. Nadeau, H. Tang, M. Snyder, Personal omics profiling reveals dynamic mo-
lecular and medical phenotypes. Cell 148,12931307 (2012).
169. A. K. Zaas, M. Chen, J. Varkey, T. Veldman, A. O. Hero III, J. Lucas, Y. Huang, R. Turner, A. Gilbert,
R. Lambkin-Williams, N. C. Øien, B. Nicholson, S. Kingsmore, L. Carin, C. W. Woods, G. S. Ginsburg,
Gene expression signatures diagnose influenza and other symptomatic respiratory viral infec-
tionsinhumans.Cell Host Microbe 6,207217 (2009).
Submitted 25 January 2013
Accepted 24 May 2013
Published 12 June 2013
10.1126/scitranslmed.3005785
Citation: J. J. McCarthy, H. L. McLeod, G. S. Ginsburg, Genomic medicine: A decade of
successes, challenges, and opportunities. Sci. Transl. Med. 5, 189sr4 (2013).
STATE OF THE ART REVIEW
www.ScienceTranslationalMedicine.org 12 June 2013 Vol 5 Issue 189 189sr4 17
... Despite promising clinical efficacy of FLT3 inhibitors, overall survival (OS) in FLT3-mutated AML vs WT is similar (77). Despite existing efforts to identify new mutations and development of targeted therapies against new targets, treatment-resistant cancers continue to emerge where non-genetic factors may also drive cancer development (78). Monotherapy targeting specific genetic abnormality is seldom effective to achieve complete response. ...
... Apart from the limited clinical benefit of genomic precision medicine, we also considered reasons that hinders the implementation of genomic precision medicine in the clinical setting. McCarthy et al. highlighted the issue of clinical validity pertaining to the use of genomic precision medicine (78). Here, clinical validity was referred as how consistent and accurate genomic testing in the clinical settings. ...
... The discovery and validation of biomarkers for each cancer type is an arduous process that requires large number of patients. The issue of identifying robust biomarker can be magnified in rare cancers with fewer number of enrolled patients (78). Another challenge with clinical implementation of GPM approach is physician's lack of knowledge in the field of genomic precision medicine (81,82) and even if they are well versed in the field of genomic precision medicine, there may still be uncertainties when interpreting genomic data (83)(84)(85) and choosing the most appropriate treatment. ...
Article
Full-text available
Acute myeloid leukemia (AML) is clinically and genetically a heterogeneous disease characterized by clonal expansion of abnormal hematopoietic progenitors. Genomic approaches to precision medicine have been implemented to direct targeted therapy for subgroups of AML patients, for instance, IDH inhibitors for IDH1/2 mutated patients, and FLT3 inhibitors with FLT3 mutated patients. While next generation sequencing for genetic mutations has improved treatment outcomes, only a fraction of AML patients benefit due to the low prevalence of actionable targets. In recent years, the adoption of newer functional technologies for quantitative phenotypic analysis and patient-derived avatar models has strengthened the potential for generalized functional precision medicine approach. However, functional approach requires robust standardization for multiple variables such as functional parameters, time of drug exposure and drug concentration for making in vitro predictions. In this review, we first summarize genomic and functional therapeutic biomarkers adopted for AML therapy, followed by challenges associated with these approaches, and finally, the future strategies to enhance the implementation of precision medicine.
... Genomic medicines due to advancements in genomics particularly led to the understanding of reasons for various rare diseases. McCarthy et al. (2013) discuss the successes, challenges, and opportunities in decoding human genomes in the last decade and present a detailed review of genomic medicines. Application of genomic data has been proved to be of utmost importance to unravel the complexities associated with rare diseases and highlighting the disease etiology and molecular mechanisms. ...
... Application of genomic data has been proved to be of utmost importance to unravel the complexities associated with rare diseases and highlighting the disease etiology and molecular mechanisms. Research studies have consistently highlighted the crucial role of genomics in understanding the genetic causes of rare diseases [7]. However, hurdles remain persistent in the clinical application of whole-genome sequencing, as pointed out by Ormond et al. (2010) [8]. ...
Research
Full-text available
This research paper studies the integration of genomic data with the methodologies of Artificial Intelligence, to create a comprehensive surveillance system that can monitor and track the prevalence of rare diseases. This research study aims to develop effective public health strategies based on data, insights, and valuable inferences from surveillance systems. This paper outlines the methodology, challenges, and potential advantages of using these two cutting edge technologies in curing and managing rare diseases.
... The advent of precision medicine and genomics marks a transformative era in healthcare, where medical treatment is tailored to the individual characteristics of each patient. This approach contrasts sharply with the "one-size-fits-all" strategy that has dominated healthcare for decades, offering a new paradigm that promises enhanced efficacy, safety, and patient outcomes (Iriart, 2019;Johnson et al., 2021;McCarthy, McLeod, & Ginsburg, 2013). At the heart of this revolution is the integration of genomics, the study of an individual's genes and their interaction with each other and the environment, into the clinical decision-making process. ...
Article
Full-text available
This review delves into Information Technology's (IT) transformative impact on precision medicine and genomics, spotlighting the pivotal role of bioinformatics, data mining, machine learning, and blockchain technologies in advancing personalized healthcare. A comprehensive analysis outlines how these IT-enabled approaches facilitate the analysis, interpretation, and application of vast genomic data sets, thereby enhancing disease prediction, diagnosis, and treatment on an individual level. Despite the promising advancements, the review also addresses significant challenges, including data complexity, interoperability, ethical considerations, and the digital divide, underscoring the necessity for multidisciplinary collaboration and innovation to overcome these hurdles. The paper concludes by emphasizing the potential of emerging technologies and patient-centred care in realizing the vision of precision medicine, which promises improved healthcare outcomes through personalized treatment strategies. Keywords: Precision Medicine, Genomics, Bioinformatics, Machine Learning, Data Security.
... Platelet index ratios, including platelet-to-lymphocyte ratio (PLR) and mean platelet volume-to-platelet count ratio (MPV/PC), have emerged as potential game-changers in the field of HIV research and treatment. [1][2][3][4][5] Traditionally associated with hemostasis and thrombosis, platelet index ratios have recently transcended their classical roles. These ratios are increasingly recognized for their ability to reflect the interplay between platelets, lymphocytes, and overall inflammatory processes within the body. ...
Article
Full-text available
Human Immunodeficiency Virus (HIV) infection is a global health challenge that requires continuous advancements in diagnostic and prognostic tools. Traditional markers, such as CD4 cell counts and viral load, have played a crucial role in monitoring disease progression and guiding therapeutic interventions. However, emerging research suggests that platelet index ratios may serve as valuable biomarkers in assessing immune health and managing HIV-associated complications. This paper explores the significance of platelet index ratios, including platelet-to-lymphocyte ratio and mean platelet volume-to-lymphocyte ratio, as potential indicators of immune system status in individuals living with HIV. The interplay between platelets, lymphocytes, and their ratios reflects the dynamic nature of the immune response and inflammatory processes during HIV infection. Understanding the role of platelet index ratios in HIV could lead to the development of accessible and cost-effective biomarkers for monitoring immune health. Implementation of these ratios in routine clinical practice may enhance the precision of disease prognosis and guide personalized treatment strategies. Additionally, the exploration of platelet index ratios may pave the way for innovative therapeutic interventions aimed at modulating immune responses in HIV-infected individuals. In conclusion, platelet index ratios represent promising emerging biomarkers for evaluating immune health and managing HIV-related complications. Further research and clinical validation are warranted to establish the utility of these ratios in routine HIV care, potentially revolutionizing the approach to monitoring and improving the health outcomes of individuals living with HIV. Abbreviations: ART = antiretroviral therapy, HIV = Human Immunodeficiency Virus, MPV = mean platelet volume, MPV/PC = mean platelet volume-to-platelet count ratio, PC = platelet count, PDW = Platelet Distribution Width, PLR = platelet-to-lymphocyte ratio.
... NGS is a high-throughput sequencing technology that sequences DNA templates along the human genome in a massively parallel manner to generate millions of DNA sequences (reads). The introduction of NGS has allowed rapid progress in many biological fields, such as clinical genetics (Krier et al., 2016;McCarthy et al., 2013), human history (Marciniak & Perry, 2017), ancient DNA analysis (Poinar et al., 2006), microbiology (Motro & Moran-Gilad, 2017), genetics (Abecasis et al., 2012), and forensic science (Palencia-Madrid & de Pancorbo, 2015;Weber-Lehmann et al., 2014). NGS technologies are routinely used to sequence whole human genomes as well as target regions to permit the sequencing of a subset of the whole genome. ...
Chapter
Forensic DNA phenotyping predicts visible physical appearance from crime scene DNA samples. Using DNA sequencing technology such as next-generation sequencing (NGS), forensic analysts can analyze hundreds of single nucleotide polymorphism (SNP) markers associated with eye, hair, and skin color to enhance the investigative power of forensic genetics. Although this technology is a milestone in forensic analysis for human identification, there are limitations on the accuracy and precision of current predictions of certain physical traits, especially those controlled by multiple genes. This chapter will explore the application of NGS technology in testing phenotype-associated SNP markers in forensics and specific SNPs linked with external physical characteristics. It will also discuss limitations, challenges, and the need for further research to enhance accuracy and precision, ultimately supporting criminal investigations.
... Thus, this has spurned efforts to transition from traditional one-size-fits-all approach to precision or personalized medicine -allowing clinicians to match the right drug to the right patient [3]. Precision medicine and targeted therapies are progressing rapidly as scientists develop a deeper understanding of patients' genome-level heterogeneity as well as prognostic and diagnostic biomarkers [4][5][6][7][8]. There are two related axes of investigation that have contributed to these efforts: pre-clinical model systems and high-throughput molecular profiling techniques; as we expound below. ...
Preprint
The pursuit of precision oncology heavily relies on large-scale genomic and pharmacological data garnered from preclinical cancer model systems such as cell lines. While cell lines are instrumental in understanding the interplay between genomic programs and drug response, it well-established that they are not fully representative of patient tumors. Development of integrative methods that can systematically assess the commonalities between patient tumors and cell-lines can help bridge this gap. To this end, we introduce the Integrative Principal Component Regression (iPCR) model which uncovers both joint and model-specific structured variations in the genomic data of cell lines and patient tumors through matrix decompositions. The extracted joint variation is then used to predict patient drug responses based on the pharmacological data from preclinical models. Moreover, the interpretability of our model allows for the identification of key driver genes and pathways associated with the treatment-specific response in patients across multiple cancers. We demonstrate that the outputs of the iPCR model can assist in inferring both model-specific and shared co-expression networks between cell lines and patients. We show that iPCR performs favorably compared to competing approaches in predicting patient drug responses, in both simulation studies and real-world applications, in addition to identifying key genomic drivers of cancer drug responses.
Chapter
This chapter embarks on an exploration of the cutting-edge developments that are reshaping the healthcare landscape. The convergence of personalized medicine and genomic research promises to revolutionize the way we approach diagnosis, treatment, and prevention of diseases. This chapter delves into the profound impact of these innovations, highlighting the shift from a one-size-fits-all approach to healthcare to one that is customized to an individual's unique genetic makeup. The authors delve into the potential of precision medicine, its applications in the treatment of diseases such as cancer, and the emerging role of genetic testing in empowering individuals to take control of their health. As we peer into the future of healthcare, we find ourselves at the threshold of a new era, where the fusion of genomics and personalized medicine promises to offer more effective, precise, and patient-centered healthcare solutions.
Article
We sympathize with many of the points Burt makes in challenging the value of genetics to advance our understanding of social science. Here, we discuss how recent reflections on epistemic validity in the behavioral sciences can further contribute to a reappraisal of the role of sociogenomics to explain and predict human traits, aptitudes, and achievement.
Article
The College of American Pathologists convened a prognostic factor conference in June 1999 to consider prognostic and predictive factors in breast, colon, and prostate cancer, and to stratify these factors into categories reflecting the strength of published evidence. Because so little progress in prognostic factor clinical utility has been made in the last 5 years, the conference participants focused their attention on decreasing variation in methods, interpretation, and reporting of these factors so that greater clarity of value could be achieved. The conference was organized to promote discussion, broad input, and future planning. An initial plenary session provided an overview of the status of tumor marker research, the impact of variation in medicine and pathology, and statistical issues related to prognostic factor research. In working group sessions for each cancer type, participants interactively evaluated and refined the documents created by the expert panels. A second plenary session dealt with issues common to all 3 groups, including the problem of micrometastases in lymph nodes in these sites; statistical issues that arose during the breakout discussions; and issues of variation in methods, interpretation, and reporting of immunohistochemical assays. A faculty session brainstormed strategies that could be used to implement the changes recommended. This session included invited representatives of the Food and Drug Administration, Health Care Financing Administration, Centers for Disease Control and Prevention, National Cancer Institute, American Joint Committee on Cancer, and International Union Against Cancer. Cancer site and general recommendations were presented and discussed during a final session to achieve consensus of the conference participants and to address feasibility of implementation of these recommendations. A final discussion focused on future initiatives that might lead to implementation of the changes proposed in the conference by the various organizations represented. This report summarizes the general conference recommendations, cancer working group recommendations, and plans for implementation of the recommendations.
Article
9500 Background: Genetic aberrations in the anaplastic lymphoma kinase (ALK) gene are found in anaplastic large cell lymphoma (ALCL), neuroblastoma (NB) and other tumors.Crizotinib,a small molecule inhibitor of ALK and c-Met, is active in non-small cell lung cancers (NSCLC) harboring an ALK translocation. We performed a phase 1 dose-escalation and pharmacokinetic (PK) trial of crizotinib in patients (pts) with refractory solid tumors and ALCL. Methods: Crizotinib was administered bid without interruption in 28 day cycles using the rolling-six design. Six dose levels (100, 130, 165, 215, 280, 365 mg/m ² /dose) have been evaluated (A1). Pts with confirmed ALK fusion proteins, mutations or amplification (A2) could enroll at one dose level lower than part A1 and those with NB could enroll on a separate stratum (A3). PK studies were performed on day 1 and at steady state (SS). ALK genomic status in tumor tissue was evaluated and qPCR was used to measure NPM-ALK fusion transcript in bone marrow and blood samples of ALCL pts. Results: 70 pts were enrolled, 57 fully evaluable for toxicity, [median (range) age 9.9 yrs (1.1–21.3)]: 29 on A1, 18 on A2, and 10 on A3. In A1, 2/7 pts developed DLT (grade 3 dizziness, grade 5 intra-tumoral hemorrhage) at 215 mg/m ² and 1/6 pts developed DLT (grade 4 liver enzyme elevation) at 365 mg/m ² . In A2, 1 grade 4 DLT (neutropenia) occurred at 165 mg/m ² ; in A3, no DLTs occurred. Mean (±SD) C ave (=AUC 0-12h /12h) of crizotinib at SS was 466±114 ng/mL at 215 mg/m ² /dose (n=5), 443±121 ng/mL at 280 mg/m ² /dose (n=8), and 720±230 ng/mL at 365mg/m ² /dose (n=4). Response data for pts with ALCL (six at 165 mg/m ² , two at 280 mg/m ² ) approved for release by the Data Safety Monitoring Committee demonstrates 7/8 (88%) complete response (CR) rate to date. RT-PCR data for 6 of these pts at 57 time points was obtained and will be described. In addition, 2 pts with NB have had CRs, one with a documented ALK mutation. One patient with an inflammatory myofibroblastic tumor and one with NSCLC had PRs. Conclusions: Inhibition of ALK in pediatric pts with ALK-driven tumors occurs with minimal toxicity and is associated with objective anti-tumor activity.
Article
Context The US Food and Drug Administration recently recommended that CYP2C19 genotyping be considered prior to prescribing clopidogrel, but the American Heart Association and American College of Cardiologists have argued evidence is insufficient to support CYP2C19 genotype testing. Objective To appraise evidence on the association of CYP2C19 genotype and clopidogrel response through systematic review and meta-analysis. Data Sources PubMed and EMBASE from their inception to October 2011. Study Selection Studies that reported clopidogrel metabolism, platelet reactivity or clinically relevant outcomes (cardiovascular disease [CVD] events and bleeding), and information on CYP2C19 genotype were included. Data Extraction We extracted information on study design, genotyping, and disease outcomes and investigated sources of bias. Results We retrieved 32 studies of 42 016 patients reporting 3545 CVD events, 579 stent thromboses, and 1413 bleeding events. Six studies were randomized trials ("effect-modification" design) and the remaining 26 reported individuals exposed to clopidogrel ("treatment-only" design). In treatment-only analysis, individuals with 1 or more CYP2C19 alleles associated with lower enzyme activity had lower levels of active clopidogrel metabolites, less platelet inhibition, lower risk of bleeding (relative risk [RR], 0.84; 95% CI, 0.75-0.94; absolute risk reduction of 5-8 events per 1000 individuals), and higher risk of CVD events (RR, 1.18; 95% CI, 1.09-1.28; absolute risk increase of 8-12 events per 1000 individuals). However, there was evidence of small-study bias (Harbord test P=.001). When analyses were restricted to studies with 200 or more events, the point estimate was attenuated (RR, 0.97; 95% CI, 0.86-1.09). In effect-modification studies, CYP2C19 genotype was not associated with modification of the effect of clopidogrel on CVD end points or bleeding (P>.05 for interaction for both). Other limitations included selective outcome reporting and potential for genotype mis-classification due to problems with the * allele nomenclature for cytochrome enzymes. Conclusion Although there was an association between the CYP2C19 genotype and clopidogrel responsiveness, overall there was no significant association of genotype with cardiovascular events. JAMA. 2011;306(24):2704-2714
Article
The HER-2/neu oncogene is a member of the crbB-like oncogene family, and is related to, but distinct from, the epidermal growth factor receptor. This gene has been shown to be amplified in human breast cancer cell lines. In the current study, alterations of the gene in 189 primary human breast cancers were investigated. HER-2/ neu was found to be amplified from 2- to greater than 20-fold in 30% of the tumors. Correlation of gene amplification with several disease parameters was evaluated. Amplification of die HER-2/neu gene was a significant predictor of both overall survival and time to relapse in patients with breast cancer. It retained its significance even when adjustments were made for other known prognostic factors. Moreover, HER-2/neu amplification had greater prognostic value than most currently used prognostic factors, including hormonal-receptor status, in lymph node-positive disease. These data indicate that this gene may play a role in the biologic behavior and/or padiogenesis of human breast cancer.