You are on page 1of 10

Clinical Chemistry 59:1

158–167 (2013) Reviews

Genomic Medicine:
New Frontiers and New Challenges
Maria D. Pasic,1,2 Sara Samaan,3,4 and George M. Yousef1,3,4*

BACKGROUND: The practice of personalized medicine applications of personalized medicine will be needed to
has made large strides since the introduction of high- address and focus on specific issues.
throughput technologies and the vast improvements © 2012 American Association for Clinical Chemistry
in computational biotechnology. The personalized-
medicine approach to cancer management holds
promise for earlier disease detection, accurate predic-
The concept of personalized medicine, which is to use
tion of prognosis, and better treatment options; how-
the individual’s genetic makeup to predict disease risk,
ever, the early experience with personalized medicine
accurately plot the course of the disease, and tailor the
has revealed important concerns that need to be ad-
management plan according to the patient’s unique
dressed before research findings can be translated to
needs, was proposed several years ago (1, 2 ). Revolu-
the bedside.
tionary improvements in the analytical tools required
for personalized medicine have accompanied the in-
CONTENT: We discuss several emerging “practical” or
troduction of high-throughput technologies. These ad-
“focused” applications of personalized medicine. Mo-
lecular testing can have an important positive impact vances, which enable simultaneous analyses of thou-
on health and disease management in a number of sands of molecules, have increased the momentum of
ways, and the list of specific applications is evolving. this field. These technologies have developed in parallel
This list includes improvements in risk assessment, with unprecedented improvements in computational
disease prevention, identification of new disease- biotechnology that allow the analysis and interpreta-
related mutations, accurate disease classification tion of the huge data sets obtained in such experiments.
based on molecular signatures, selection of patients Finally, the introduction of targeted therapy has in-
for enrollment in clinical trials, and development creased the demand for molecular characterization of
of new targeted therapies, especially for metastatic diseases to identify new candidate markers for progno-
tumors that are refractory to treatment. Several sis and prediction (3–5 ) and to facilitate the develop-
challenges remain to be addressed before genomics ment of new targeted therapies (6 – 8 ).
information can be applied successfully in the rou- Such molecular analyses are predicted to be im-
tine clinical management of cancers. Further im- portant for managing cancer patients by offering im-
provements and investigations are needed in data provements in early cancer detection, accurate predic-
interpretation, extraction of actionable items, cost- tion of prognosis, and better treatment options with
effectiveness, how to account for patient heterogeneity minimal side effects (1, 9, 10 ). Information gained by
and ethnic variation, and how to handle the risk of these methods will also help in evolving our under-
“incidental findings” in genetic testing. standing of disease pathogenesis by switching the focus
to a search for altered “biological processes” from a
SUMMARY: It is now clear that personalized medicine focus on the discovery of individual disease biomarkers
will not immediately provide a permanent solution for (4 ). The change to processes rather than markers will
patient management and that further refinement in the be useful for investigating complex diseases, such as
cancer, diabetes, and hypertension, which often in-
volve large numbers of genes, pathways, and environ-
mental factors (4, 5 ). Thus, we are stepping into a new
1
Department of Laboratory Medicine and Pathobiology, University of Toronto, era of “genomic” medicine. It is not surprising that
Toronto, Ontario, Canada; 2 current affiliation: Department of Laboratory Med-
icine, St. Joseph’s Health Centre, Toronto, Ontario, Canada; 3 Department of 10% of the labels for drugs cleared by the U.S. Food and
Laboratory Medicine, St. Michael’s Hospital, Toronto, Ontario, Canada;
4
Drug Administration already contain pharmacog-
Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s
Hospital, Toronto, Ontario, Canada.
enomic information (11 ), and this number will only
* Address correspondence to this author at: Department of Laboratory Medicine, increase as genomic testing becomes increasingly used
St. Michael’s Hospital, 30 Bond St., Toronto, Ontario, M5B 1W8 Canada. Fax in routine care (12 ).
416-864-5648; e-mail yousefg@smh.ca.
Received September 11, 2012; accepted October 24, 2012. After a period of initial enthusiasm for personal-
Previously published online at DOI: 10.1373/clinchem.2012.184622 ized medicine, it became apparent that several impor-

158
Genomic Medicine in Cancer
Reviews

Table 1. The potential applications of genomic Table 2. Strategies to increase the cost-
medicine. effectiveness of genomic screening strategies.

1. Examination of variation among healthy individuals 1. Target screening to a high-risk population


2. Understanding disease risk, susceptibility, and etiology 2. Focus on chronic diseases
3. Disease prevention 3. Focus screening on diseases with known interventions
4. Early diagnosis at the preclinical stage (to help modify 4. Identify molecular changes directly related to short-term
disease course, e.g., diet and exercise in diabetes) benefits
5. Identification of new disease-related mutations 5. Limit screening to diseases with a higher prevalence
6. Accurate diagnosis of challenging cases with inconclusive 6. Interpret data in a clinical context (e.g., environmental
results for clinical parameters factors)
7. Accurate disease classification based on molecular signature
8. Health management (prognosis and predictive markers)
9. Identification of personal drug-related profile
ment of a recognized drug response, and pathogenicity
(14 ). This study also highlights the importance of using
10. Selection of patients for clinical trials
the expanding number of disease-specific mutation
11. Monitoring disease status (e.g., recurrence) and pharmacogenomics databases as valuable sources
12. Monitoring tumor evolution in response to treatment for identifying disease risk.
13. Developing new targeted therapies, especially for metastatic Moreover, an interesting study has demonstrated
tumors refractory to treatment the potential power of exome sequencing to render a
“molecular-based diagnosis.” A novel mutation was
identified in a patient with Crohn disease after all stan-
tant concerns needed to be addressed before these dis- dard diagnostic modalities had failed to reach the cor-
coveries can be translated to the bedside (13 ). It is now rect etiologic diagnosis (15 ). This and similar reports
clear that personalized medicine is not going to imme- have shown the feasibility of using comprehensive
diately provide a permanent solution for all patient- genomic analyses for identifying causes of known dis-
management issues and that we need to refine its ap- eases. These studies have underscored the need to ad-
plications for specific and focused issues. In this review, dress many of the issues related to implementing
we discuss some of the emerging “practical” or “fo- genomic testing directly into the clinic, as we detail
cused” applications of personalized medicine. In addi- below.
tion, we use cancer as an example for highlighting the A pressing question to be addressed is how to esti-
challenges to be faced as we step into the era of person- mate the accuracy of sequencing technologies for pre-
alized medicine. dicting disease risk. Kohane et al. recently provided an
excellent review on this topic (16 ). Roberts et al. ana-
Evolution of New Applications in Patient lyzed the capability of personal genome sequencing for
Management predicting the risk of 24 different diseases (17 ). On the
negative side, these investigators found that most mo-
Table 1 presents an evolving list of specific applications nozygotic twins had negative results in sequencing tests
in which molecular testing can play an important role for 23 of the 24 diseases. On the positive side, however,
in improving health and disease-management plans. about 95% of the tested individuals were alerted to
clinically important predispositions for at least 1 dis-
THE USE OF GENOME SEQUENCING AND INTEGRATED ease. A related major challenge is the ability to reliably
MOLECULAR ANALYSIS FOR ASSESSING DISEASE RISK distinguish pathogenic mutations, benign variants,
Next-generation sequencing has several promising and variants of uncertain importance. Moreover, anal-
applications in personalized medicine. Predicting yses of diseases with a complex, multifactorial etiology,
risk through molecular analysis, usually via next- such as hypertrophic cardiomyopathy, which has a vast
generation sequencing, can have an important impact genetic variability and a high frequency of novel muta-
on disease outcomes. Genome sequencing also can tions, have revealed unforeseen difficulties in translat-
make important contributions to reproductive health. ing complex science into the clinical arena.
Such improvements include prescreening of mothers Other major concerns in this regard are the ability
for mutations related to metabolic and other mende- to translate this information into meaningful health
lian disorders (14 ). A recent study showed the ability of improvements and the costs of these expensive screen-
an integrated analysis to identify genetic variants asso- ing tools. Table 2 lists several strategies for improving
ciated with the risks of mendelian diseases, develop- the cost-effectiveness of genomic screening. One strat-

Clinical Chemistry 59:1 (2013) 159


Reviews

egy is to restrict the use of molecular screening tests for ing evidence suggests that integrating multiple levels of
identifying disease susceptibility to individuals in molecular data holds great promise for refining our
“clinically appropriate” categories, e.g., populations at understanding of disease pathogenesis and patient
risk (18 ). Furthermore, assessing risks for such chronic management (14, 27, 28 ). Appropriate data collection
diseases as diabetes, hypertension, and cancer is eco- and database integration will be necessary to efficiently
nomically efficient, because it will substantially reduce assess the potential clinical and biological importance
the burdens of treatment and follow-up for extended of rare genetic variants (1, 29 ).
periods of time. It is also advantageous to focus on Moreover, now becoming available are several na-
identifying the diseases with known interventions that tional and international public databases that integrate
can modify or alleviate disease progression (18 ). An- different levels of molecular high-throughput analyses
other practical approach is to look for molecular to draw meaningful “actionable” conclusions that can
changes that are directly related to short-term benefits help in managing patients. Examples of such databases
for managing patient outcomes. Several genomic stud- are i2b2 (Informatics for Integrating Biology & the
ies have tried to identify patients with the greatest like- Bedside; https://www.i2b2.org) (30 ) and locus-specific
lihood of receiving short-term benefits from whole- mutation databases, such as the Human Gene Muta-
genome sequencing tests, specifically in the areas of tion Database, or HGMD (http://www.hgmd.cf.ac.uk/
diagnosis and management (19 –21 ). ac/index.php) (31 ). Several databases also catalog so-
matic mutations occurring in different cancers (32 ).
INTEGRATED GENOMICS DATABASES The promising preliminary analyses of such data have
Several genomewide association studies have applied shown that many of these mutations are present in
analyses of single-nucleotide polymorphisms (SNPs)5 many types of cancer and that they are targetable via
to assess for the presence of genetic variants in different drug therapy (29 ).
individuals and their potential associations with dis- Integrated analyses are best interpreted in the clin-
ease risk. The first successful study, published in 2005, ical contexts of a specific patient, such as medical his-
investigated age-related macular degeneration (22 ). tory, family history, physical/clinical assessments, im-
Since then, thousands of genomewide association stud- munological factors for risk prediction, and so forth
ies of humans have investigated ⬎200 diseases and (33 ). Also important is to recognize that molecular
traits, and almost 4000 SNP associations have been changes are the product of both genomic alterations
identified (23 ). On the oncology frontier, several inter- and the environment (food, exercise, and so forth) and
national projects have been developed to catalog mul- that some mutations and SNPs are reflections of envi-
tilevel somatic alterations in cancer through analyses of ronmental changes. Therefore, molecular data ob-
exome sequences, DNA copy numbers, promoter tained via high-throughput technologies must be
methylation, and mRNA and microRNA (miRNA) interpreted in the context of clinical variables and en-
production. These projects include the Cancer Ge- vironmental conditions to provide a better assessment
nome Atlas (http://cancergenome.nih.gov/) (24 ), the of a patient’s risk of disease.
Cancer Genome Project (25, 26 ), and the International
Cancer Genome Consortium (25 ). Furthermore, to al- MONITORING THE PERSONAL GENOME
low broad analyses of genomic variation in several dis- A new, promising approach is “integrative personal omics
ease conditions, the NIH has launched various initia- profiling” (iPOP) (34 ). iPOP combines genomic, tran-
tives, including providing support for genome analyses scriptomic, proteomic, metabolomic, and autoantibody
of participants in the Framingham Heart Study, col- profiles from the same individual to follow their genomic/
lecting samples from infants in the National Chil- transcriptomic composition over long periods— during
dren’s Study, and conducting genetic analyses of 20 times of health and disease. A recent study demon-
different types of tumors (2 ). Other informative ini- strated that longitudinal iPOP can evaluate healthy
tiatives include cataloging the variation among and disease states by connecting the genomic infor-
healthy individuals, such as the 1000 Genomes proj- mation with additional dynamic “omics” activities
ect (http://www.1000genomes.org/). (34 ). Chen et al. repeatedly collected data from a
Also evolving are new databases that incorporate single individual over time. The data analysis re-
multilevel molecular information for assessing disease vealed heteroallelic changes in both the healthy and
risk and responses to different drugs. The accumulat- disease states, as well as changes in RNA-editing
mechanisms, that would have been masked other-
wise. The results indicated that differential allele-
specific expression is extensive in humans, with es-
5
Nonstandard abbreviations: SNP, single-nucleotide polymorphism; miRNA, pecially distinct differences between healthy and
microRNA; iPOP, integrative personal omics profiling. disease states (34 ). This approach allowed the inte-

160 Clinical Chemistry 59:1 (2013)


Genomic Medicine in Cancer
Reviews

gration of multiple profiles associated with different forms include the MiSeq Personal Sequencer and
physiological states across multiple time points, HiSeq platforms (Illumina) and semiconductor se-
thereby providing detailed information that, owing quencing (Ion Torrent Systems/Life Technologies).
to interindividual variability, would not have been
apparent in group studies (34 ). Furthermore, a large TARGETED THERAPY AND DRUG TRIALS
database can be created with complete profiles from Molecular information can also be used for improving
more individuals with different types of diseases. treatment, either through development of targeted
Such a database may be valuable in the diagnosis, therapy or rediscovery of previously failed drugs for use
monitoring, and treatment of diseases (34 ). in subgroups of patients likely to benefit from them
(2 ). The numbers remain small, however, both for
DISEASE DIAGNOSIS AND BIOLOGICAL CLASSIFICATIONS known “actionable” genetic abnormalities and for the
Molecular signatures can be the basis for discovering targeted agents that are to be tested within the next few
new diagnostic markers for many diseases. There is a years (1 ).
wealth of literature on the use of differential gene ex- Molecular markers can also be used to predict
pression and protein production for identifying new treatment efficacy so that patients not expected to re-
biomarkers for early disease detection and assessment spond to therapy will avoid unnecessary treatments.
of prognosis (35, 36 ). Furthermore, molecular markers can serve as prognos-
As mentioned above, exome sequencing has iden- tic markers of disease aggressiveness, information that
tified a novel mutation that permitted a molecular- could be reflected in treatment plans. An interesting
based diagnosis for a patient with Crohn disease. On example is the concern regarding “overtreatment” of
another interesting frontier, genomic analyses hold prostate cancer patients who have less aggressive forms
promise for establishing new biological subclassifica- of the cancer and would better be candidates for “active
tions of many diseases, including cancer, which could surveillance.” Such patients are awaiting the develop-
change how cancer is managed. Molecular signatures are ment of accurate molecular markers that can better
now used to classify tumors according to their “biological guide their treatment choices (44 ). Well-designed pro-
behavior,” rather than by their tissue, organ of origin, or spective clinical trials are needed to measure patient
morphology (37 ). Examples of molecular signatures are outcomes for various genomic applications, the advan-
documented in the literature. DNA microarray data have tages of which are not yet evident compared with cur-
permitted the subcategorization of lymphomas and glio- rent standards of care (1, 12 ).
blastomas (38 ), and alternative-splicing data obtained Molecular signatures are also expected to revolu-
with microarrays have aided in defining breast cancer tionize the eligibility criteria for clinical trials. Cur-
subtypes and improving treatment options (39 ). A recent rently, inclusion criteria for clinical trials are organ
study has documented that data on the differential pro- based. The availability of molecular data allows the
duction of miRNAs are capable of accurately classifying possibility of switching to pathway-based selection cri-
the different subtypes of kidney cancer (40 ). Another teria for clinical trial enrollment, rather than organ-
study has highlighted the ability of unique molecular pro- based criteria (37 ). Inclusion criteria for such trials
files to accurately predict prostate cancer prognosis, espe- should allow patients with different tumor types to en-
cially in patients with low or average Gleason scores (41 ), roll in a study as long as they have the biological targets
and miRNA production signatures have accurately pre- and mutations relevant to the therapy being evaluated.
dicted the risk of biochemical failure in prostate cancer at A recent study has shown that integrative high-
the time of surgery (42 ). In addition, a growing body of throughput sequencing of patients with cancers in
evidence supports the potential utility of genotype advanced stages can generate a comprehensive muta-
changes for predicting clinical outcome and response to tional landscape that would facilitate biomarker-
treatment (43 ). driven clinical trials in oncology (28 ).
An important step toward the translation of these Several web-based applications have recently been
applications to the clinic is the advent of commercially established to identify and catalog mutations that have
available systems for screening large numbers of muta- therapeutic implications and can affect patient enroll-
tions simultaneously. These systems include Mass- ment in clinical trials. One such endeavor is My Cancer
ARRAY (Sequenom; http://www.sequenom.com), Genome (http://www.mycancergenome.com) (45 ).
which can be used with predesigned, optimized panels Another interesting area to explore is to apply
(such as the OncoCarta™ panel) or custom-designed genomic approaches to pharmacogenomics. Genomic
arrays. Another system, the IonAmpliSeq™ Cancer analysis can play an important role in predicting treat-
Panel (Invitrogen/Life Technologies), can simultane- ment efficacy for individual patients (46 ). To appreci-
ously analyze 739 COSMIC (Catalogue of Somatic Mu- ate the importance of pharmacogenomics, consider
tations in Cancer) mutations at 604 loci. Similar plat- that ⬎2 ⫻ 106 adverse drug reactions occur annually in

Clinical Chemistry 59:1 (2013) 161


Reviews

the US alone. The approximately 1000 deaths that re-


sult make adverse drug reactions the fourth leading Table 3. Potential challenges of the era of genomic
cause of death (47 ). Moreover, the estimated annual medicine.
cost of drug-related morbidity and mortality in the US
is ⬎$76 billion. Added to this is the waste of medica- 1. Data interpretation and extraction of actionable items
tions administered to the patients who do not respond. 2. Guidelines for implementing new molecular testing
On average, a given medication will have a therapeutic 3. Cost-effectiveness of molecular testing
effect in only 25% to 60% of patients (48 –50 ). 4. Patient heterogeneity and ethnic variation
Treatments guided by pharmacogenomic evalua- 5. Test optimization and standardization
tions will also overcome an important limitation of
6. The risk of incidental findings and false-positive results
current treatment strategies, which base drug efficacy
7. Need for training and teamwork efforts
on the average response in clinical trials, not the re-
sponses of the individual patients. The reality is that 8. Between-platform variation in results
responses to treatment vary greatly among individuals;
thus, the optimal approach is to determine targeted interactions, the goal of which is to gain a better under-
therapies individually, in accordance with the unique standing of diseases with complex etiology, such as in-
genetic composition of each patient (46 ). flammatory diseases, in which microbial elements play
a role in pathogenesis (58 ). On the one hand, metag-
THE ROLE OF PROTEOMICS IN GENOMIC MEDICINE enomics analyses may facilitate a much better under-
Currently, most techniques for personalized medicine are standing of pathogenesis at the pathway level of analy-
based on genomics; however, proteomic techniques— sis (59 ). On the other hand, however, extracting
including measurement of individual circulating or meaningful information from these multilevel analyses
tissue-based proteins, identification of disease-specific is becoming more difficult. There is a need to focus on
posttranslational modifications, and generation of developing interpretation pipelines for omics investi-
prognostic/predictive protein signatures for fluids or gations that identify “actionable” or “druggable” items
tissues—are increasingly being used to assist in diagno- that can directly improve patient outcomes and that
sis, prognosis, therapy selection, and predicting thera- can connect all the steps between the identification of a
peutic responses to drugs (25, 51–53 ). Historical pro- potential therapeutic target in the laboratory setting
teomic tests, such as measuring estrogen and and the approval of a therapy for clinical use (i.e., mov-
progesterone receptor proteins in breast tumor tissues ing candidate compounds into commercial develop-
to predict a patient’s response to tamoxifen, are still ment) (60 ). An interesting strategy that also shows
widely used. Recently, mass spectrometry has been promise is the classification of molecular changes into
used to identify pancreatic cancer proteomes. These those that represent the “primary etiology” and those
efforts have led to the identification of new candidate that are “contributing modifiers,” because, ultimately,
biomarkers, including at least 1 marker, CUZD1 (CUB not all that glitters is gold.
and zona pellucida–like domain-containing 1), that By the time a cancer is diagnosed, billions of cells
has a sensitivity and a specificity superior to the cur- containing gene abnormalities have already been pro-
rently used cancer antigen marker CA19-9 (54 ). Others duced, and many secondary genetic mutations are be-
have used these techniques to identify markers of tax- ing acquired. Some of these secondary mutations may
ane sensitivity in breast cancer patients receiving adju- be drivers (mutations selected for during tumorigene-
vant paclitaxel and radiation therapy (55 ) and to iden- sis), and some may be passengers (incidental or effector
tify new prognostic biomarkers (56 ). mutations). Mutations in both classes arise via such
We anticipate that we will soon be able to quantify processes as mutational exposures, genome instability,
thousands of proteins and assess the function or disease and large numbers of cell divisions (61 ). Driver genes
of many organs simply by using a drop of blood (57 ). are ideal candidates for actionable targets.
The goal of delivering genetic testing to patients
New Challenges for Genomic Medicine faces several challenges, including determining which
genetic markers have the most clinical importance,
We are witnessing a revolutionary era with a large limiting the off-target effects of gene-based therapies,
number of omics, including genomics (chromosomes, conducting clinical trials to identify genetic variants
SNPs), epigenetics (methylation, miRNA), transcrip- that correlate with drug response, and regulating ge-
tomics (mRNA, splice variants), proteomics, cyto- netic testing to protect patients while still encouraging
kines, metabolomics, antibody-omics, and micro- innovation (2 ). Table 3 is a partial list of some of the
biomics. Another interesting emerging field is potential challenges to be addressed. Some of these
“metagenomics,” the study of host–microbe genomic challenges have recently been addressed in detail (25 ).

162 Clinical Chemistry 59:1 (2013)


Genomic Medicine in Cancer
Reviews

DEVELOPING AND IMPLEMENTING NEW MOLECULAR TESTS necessary to make the results of all studies publicly
A challenge that should be considered is the setting of available and easily accessible.
rules for implementing new molecular testing in the Another important step to be addressed before
clinical laboratory. Currently, the trigger for molecular personalized genomics can move into clinical applica-
testing can come from the treating physician in re- tions is the consolidation of multidisciplinary teams of
sponse to information obtained from a recent journal clinicians, laboratory scientists, and bioinformaticians
article or at a meeting. The trigger can also be a new who can extract meaningful information from the mil-
program initiative at the institution. A substantial pro- lions of data points regarding molecular changes.
portion of new molecular testing is driven by the pa- Accompanying the evolution of applications in
tient in response to the electronic media. A well- genomic medicine is the need to provide caregivers in-
structured committee should be in place to evaluate formation about these new applications. Understand-
new testing on the basis of cost-effectiveness and the ing the benefits and challenges of molecular testing is a
higher levels of confidence that evidence-based medi- key issue that will have to be addressed. Genetic tests
cine requires (62, 63 ). are not perfect because most gene mutations do not
Among the technical challenges that should be predict outcomes perfectly. Thus, clinicians will need
considered when developing a new test is the variation to understand the specificity and sensitivity of new di-
between different platforms that measure the same agnostic measures (2, 12 ). It will also be necessary to
molecular changes. A recent study of 7 diseases dem- educate clinicians about whether or when to order cer-
onstrated an agreement of ⱕ50% between the 2 plat- tain diagnostic tests. For instance, it would be inappro-
forms evaluated (64 ). priate to order a test when the clinician is unsure about
Cost-effectiveness remains an important concern how to use the results [e.g., for rare sequence variants in
in implementing new molecular tests. In this regard, BRCA16 (breast cancer 1, early onset) and BRCA2
molecular testing of markers predictive for targeted (breast cancer 2, early onset) genes in a breast cancer
patient] (12 ). In addition, the feedback between clini-
therapy in metastatic cancer represents an ideal model
cal practice and data interpretation is also an important
for application, because patients with metastatic cancer
factor to consider. The information that clinicians can
undergo costly drug therapies that have serious adverse
give to biochemists and bioinformaticians is poten-
reactions (65 ). Although a genomics-based predictive
tially crucial.
test can be expensive, it may eventually lead to substan-
Equally important is the need to train the new gen-
tial financial savings by excluding a large proportion of
eration of clinicians and laboratory scientists to under-
patients predicted to be nonresponders. In addition, it stand the different dimensions of these evolving
would substantially reduce the costs of treating side applications. Training programs, such as the TRIG
effects and treatment complications. (Training Residents in Genomics) curriculum (http://
It must also be recognized that developing a new www.pathologytraining.org), have already been estab-
test is a complicated multistep process that continues lished. Studies have suggested that the “genetic liter-
after the discovery phase. Validation, especially inde- acy” of the general public is inadequate to prepare
pendent validation, is one of the necessary steps to be citizens for this unprecedented access to genomic in-
completed; others steps include standardization, test formation. Given that the current generation of high
interpretation, the technical range, quality control, and school students will come of age in an era when per-
so on (66, 67 ). sonal genetic information is becoming increasingly
Another important consideration is that “test ac- used in healthcare, ensuring that students understand
curacy” improves with time; consequently, reevaluat- the genetic concepts necessary to make informed med-
ing test performance might be necessary. An example ical decisions is of vital importance (68 ).
of this consideration is hormone receptor testing in
breast cancer, in which improvements in sensitivity THE RISK OF THE INCIDENTALOME IN GENETIC TESTING
and specificity for the antibody used and additional As the number of combined tests increases, the false-
evidence on hormone treatment efficacy prompted a discovery rate will become very high, perhaps reaching
change in the cutoff for positivity. 60% if 10 000 SNPs are analyzed simultaneously. This
increased false-positive rate represents a phenomenon
THE NEED FOR TRAINING AND TEAMWORK EFFORTS known as the “incidentalome,” a concatenation of a
In the era of personalized genomics, it is important to
emphasize the concept of collaboration rather than
competition. Making the huge costs of such projects
economically feasible requires different centers world- 6
Human genes: BRCA1, breast cancer 1, early onset; BRCA2, breast cancer 2,
wide to work together. International legislation will be early onset.

Clinical Chemistry 59:1 (2013) 163


Reviews

Fig. 1. Schematic showing the multistep process of translating genomics into the clinical setting.
Multilevel molecular analysis should be integrated and interpreted by a multidisciplinary team that will decide on the proper
clinical implementation. Genomic markers can be used to assess disease risk or serve as diagnostic or prognostic markers; they
can also have therapeutic applications. aCGH, array-comparative genomic hybridization.

huge number of complex positive findings that are “real” risks associated with positive results in screening
poorly understood (16 ). tests and to help physicians make cost-effective deci-
Kohane et al. highlighted the fact that substantially sions. We obviously have a long way to go before we
increasing the number of tested molecules will produce truly understand the relationship between the millions
substantial increases in the numbera of false-positive of SNPs and disease risk. Interestingly, even SNPs that
results, especially in a population with a low disease have a documented effect on the population with dis-
prevalence (16 ). That will create several problems, in- ease might not confer a risk for the same effect in an
cluding unnecessary and costly follow-up studies for asymptomatic general population (69 ).
falsely identified diseases. Such studies will add to the
already burdened healthcare system. PATIENT HETEROGENEITY AND ETHNIC VARIATION
The lack of accuracy and precision for screening Ethnic variation should be considered when interpret-
tests will put physicians into the challenging situation ing genomic data and should be carefully addressed
of either ignoring the results of incidental findings when translating results of high-throughput genetic
(with the subsequent risk of liability) or expending analyses to the clinic. Another important challenge is
more resources to perform confirmatory “gold stan- the heterogeneity that exists among patients with the
dard” testing. Several steps have been suggested to re- same cancer type. Recent studies have shown that only
duce the impact of the incidentalome. They include a small number of gene mutations (also called “moun-
assessing the overall disease prevalence and using in- tains”) are present in the majority of a large population
formation system technologies to understand the of patients with a particular cancer, whereas most other

164 Clinical Chemistry 59:1 (2013)


Genomic Medicine in Cancer
Reviews

genes (known as “hills”) are mutated less frequently. consumer genetic testing should be evaluated cautiously
Studies of cancer genome landscapes have found gene in terms of its reliability and the impact of these tests on
“mountains” in a large proportion of tumors and gene the public. In addition, providing these tests to the public
“hills” in ⬍5% of tumors (70 ); however, most muta- must be regulated (72 ).
tions in cancers that are likely to be driver mutations do
not occur in mountains. Ultimately, the hills drive tu- Concluding Remarks
mor progression (70 ). The landscape of individual pa-
tients has also been shown to overlap very little with the Despite the many existing challenges, there is light at
overall landscape of consensus mutations. This fact ac- the end of the tunnel. The cost of high-throughput
counts for the great heterogeneity in biomarker expres- analyses has dramatically declined. For instance, $1000
sion observed among cancer patients (70 ). Adding to full-genome sequencing will become a reality in the
the complexity of the problem is the fact that not all very near future. Additionally, targeted approaches
driver mutations that occur with tumor progression that focus on specific “actionable” molecules will bring
are essential for tumor maintenance; consequently, down the costs of molecular testing to a very reasonable
they may not be good therapeutic targets (61 ). The level (28 ). Furthermore, the costs of whole-genome se-
ability to identify driver mutations and to distinguish quencing will be small when they are amortized over an
them from the many incidental mutations will help in individual’s expected lifetime, because the baseline
capturing the full genetic heterogeneity in cancer, a feat genomic information can be consulted at any stage of
that will require a very large sample size (61 ). life. Finally, as the cost of biomarker discovery de-
Another interesting obstacle is the presence of in- creases, the rate of discovery will accelerate (12 ).
tratumoral heterogeneity. A recent elegant study has We are indeed moving into a new revolutionary
shown that analyzing a single sample obtained from a era of personalized medicine. The recognition of the
single region of a tumor can underestimate intratu- merits and limitations of genomic medicine can guide
moral heterogeneity (i.e., the tumor’s genomic land- us to more-realistic expectations and foster new direc-
scape). Such heterogeneity may present a big challenge tions for research (74 ). At present, we can generate
for biomarker-identification approaches. It can also af- data much faster than we can interpret them. Fig. 1 is a
fect therapeutic choices (71 ). proposed outline of the multistep process required to
achieve “real-life” applications of high-throughput
ETHICAL AND LEGAL CONSIDERATIONS
genomic data. The contribution of genomic analysis is
Several ethical considerations should be carefully ad- no longer controversial (69 ), and genomic data, com-
dressed. They include the accessibility of test results, espe- bined with transcriptomic, proteomic, and metabolo-
cially for other family members who might be at risk for mic data, are expected to provide a much deeper un-
the same disease, prospective employers, and legal au- derstanding of the healthy and disease states (10 ).
thorities (72 ). Another interesting point is the reporting
of incidental findings. Should the patients be informed of
only results that are directly related to the disease of con-
cern, or should all results, including incidental findings,
Author Contributions: All authors confirmed they have contributed to
be reported? There are also concerns about reporting the
the intellectual content of this paper and have met the following 3 re-
potential risk of developing a disease for which no pro- quirements: (a) significant contributions to the conception and design,
phylactic measures exist and whether such reporting acquisition of data, or analysis and interpretation of data; (b) drafting
would lead to anxiety in the patient. The issue of securing or revising the article for intellectual content; and (c) final approval of
these large amounts of data is also a concern (73 ). Patent- the published article.
ing issues (Who owns the genes?) is another ongoing de- Authors’ Disclosures or Potential Conflicts of Interest: No authors
bate in the scientific community. Finally, direct-to- declared any potential conflicts of interest.

References
1. Dancey J. Genomics, personalized medicine and 5. La Thangue NB, Kerr DJ. Predictive biomarkers: a 8. Gonzalez-Angulo AM, Hennessy BT, Mills GB.
cancer practice. Clin Biochem 2012;45:379 – 81. paradigm shift towards personalized cancer medi- Future of personalized medicine in oncology: a
2. Hamburg MA, Collins FS. The path to personal- cine. Nat Rev Clin Oncol 2011;8:587–96. systems biology approach. J Clin Oncol 2010;28:
ized medicine. N Engl J Med 2010;363:301– 4. 6. Galvan A, Skorpen F, Klepstad P, Knudsen AK, 2777– 83.
3. Abrahams E. Right drug—right patient—right Fladvad T, Falvella FS, et al. Multiple loci modu- 9. Metias SM, Lianidou E, Yousef GM. MicroRNAs in
time: Personalized Medicine Coalition. Clin Transl late opioid therapy response for cancer pain. Clin clinical oncology: at the crossroads between
Sci 2008;1:11–2. Cancer Res 2011;17:4581–7. promises and problems. J Clin Pathol 2009;62:
4. Kelloff GJ, Sigman CC. Cancer biomarkers: select- 7. Gazdar AF. Personalized medicine and inhibition 771– 6.
ing the right drug for the right patient. Nat Rev of EGFR signaling in lung cancer. N Engl J Med 10. Snyder M, Du J, Gerstein M. Personal genome
Drug Discov 2012;11:201–14. 2009;361:1018 –20. sequencing: current approaches and challenges.

Clinical Chemistry 59:1 (2013) 165


Reviews

Genes Dev 2010;24:423–31. large-scale molecular data sets. Nucleic Acids Res mesylate for treatment of advanced gastrointes-
11. Frueh FW, Amur S, Mummaneni P, Epstein RS, 2012;40:D109 –14. tinal stromal tumor: CALGB 150105 Study by
Aubert RE, DeLuca TM, et al. Pharmacogenomic 28. Roychowdhury S, Iyer MK, Robinson DR, Lonigro Cancer and Leukemia Group B and Southwest
biomarker information in drug labels approved by RJ, Wu YM, Cao X, et al. Personalized oncology Oncology Group. J Clin Oncol 2008;26:5360 –7.
the United States Food and Drug Administration: through integrative high-throughput sequencing: 44. Cooperberg MR, Carroll PR, Klotz L. Active sur-
prevalence of related drug use. Pharmacotherapy a pilot study. Sci Transl Med 2011;3:111ra121. veillance for prostate cancer: progress and prom-
2008;28:992– 8. 29. Dancey JE, Bedard PL, Onetto N, Hudson TJ. The ise. J Clin Oncol 2011;29:3669 –76.
12. Feero WG, Guttmacher AE, Collins FS. Genomic genetic basis for cancer treatment decisions. Cell 45. Web applications aid clinical trial recruitment.
medicine—an updated primer. N Engl J Med 2012;148:409 –20. Cancer Discov 2012;2:OF2.
2010;362:2001–11. 30. Natter MD, Quan J, Ortiz DM, Bousvaros A, 46. Ginsburg GS, Willard HF. Genomic and personal-
13. Chan IS, Ginsburg GS. Personalized medicine: Ilowite NT, Inman CJ, et al. An i2b2-based, gen- ized medicine: foundations and applications.
progress and promise. Annu Rev Genomics Hum eralizable, open source, self-scaling chronic dis- Transl Res 2009;154:277– 87.
Genet 2011;12:217– 44. ease registry. J Am Med Inform Assoc 2013;20: 47. Severino G, Del ZM. Adverse drug reactions: role
14. Ashley EA, Butte AJ, Wheeler MT, Chen R, Klein 172–9. of pharmacogenomics. Pharmacol Res 2004;49:
TE, Dewey FE, et al. Clinical assessment incorpo- 31. Stenson PD, Ball EV, Howells K, Phillips AD, Mort 363–73.
rating a personal genome. Lancet 2010;375: M, Cooper DN. The Human Gene Mutation 48. Deverka PA, Vernon J, McLeod HL. Economic
1525–35. Database: providing a comprehensive central mu- opportunities and challenges for pharmacog-
15. Worthey EA, Mayer AN, Syverson GD, Helbling D, tation database for molecular diagnostics and enomics. Annu Rev Pharmacol Toxicol 2010;50:
Bonacci BB, Decker B, et al. Making a definitive personalized genomics. Hum Genomics 2009;4: 423–37.
diagnosis: successful clinical application of whole 69 –72. 49. Tsongalis GJ, Ricci A Jr. Breast cancer as a model
exome sequencing in a child with intractable 32. Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, of realistic challenges in pharmacogenomics. Clin
inflammatory bowel disease. Genet Med 2011; Beare D, et al. COSMIC: mining complete cancer Biochem 2003;36:89 –94.
13:255– 62. genomes in the Catalogue of Somatic Mutations 50. Zhou SF, Di YM, Chan E, Du YM, Chow VD, Xue
16. Kohane IS, Masys DR, Altman RB. The in Cancer. Nucleic Acids Res 2011;39:D945–50. CC, et al. Clinical pharmacogenetics and potential
incidentalome: a threat to genomic medicine. 33. Raes J, Letunic I, Yamada T, Jensen LJ, Bork P. application in personalized medicine. Curr Drug
JAMA 2006;296:212–5. Toward molecular trait-based ecology through Metab 2008;9:738 – 84.
17. Roberts NJ, Vogelstein JT, Parmigiani G, Kinzler integration of biogeochemical, geographical and 51. Piran S, Liu P, Morales A, Hershberger RE. Where
KW, Vogelstein B, Velculescu VE. The predictive metagenomic data. Mol Syst Biol 2011;7:473. genome meets phenome: rationale for integrat-
capacity of personal genome sequencing. Sci 34. Chen R, Mias GI, Li-Pook-Than J, Jiang L, Lam HY, ing genetic and protein biomarkers in the diag-
Transl Med 2012;4:133ra58. Chen R, et al. Personal omics profiling reveals nosis and management of dilated cardiomyopa-
18. Khoury MJ, McCabe LL, McCabe ER. Population dynamic molecular and medical phenotypes. Cell thy and heart failure. J Am Coll Cardiol 2012;60:
screening in the age of genomic medicine. N Engl 2012;148:1293–307. 283–9.
J Med 2003;348:50 – 8. 35. Siu KW, DeSouza LV, Scorilas A, Romaschin AD, 52. Carethers JM. Proteomics, genomics, and molec-
19. Boraska V, Davis OS, Cherkas LF, Helder SG, Honey RJ, Stewart R, et al. Differential protein ular biology in the personalized treatment of
Harris J, Krug I, et al. Genome-wide association expressions in renal cell carcinoma: new bio- colorectal cancer. J Gastrointest Surg 2012;16:
analysis of eating disorder-related symptoms, be- marker discovery by mass spectrometry. J Pro- 1648 –50.
haviors, and personality traits. Am J Med Genet B teome Res 2009;8:3797– 807. 53. Kuzmanov U, Smith CR, Batruch I, Soosaipillai A,
Neuropsychiatr Genet 2012;159B:803–11. 36. White NM, Bao TT, Grigull J, Youssef YM, Girgis Diamandis A, Diamandis EP. Separation of kal-
20. Lui JC, Nilsson O, Chan Y, Palmer CD, Andrade A, Diamandis M, et al. miRNA profiling for clear likrein 6 glycoprotein subpopulations in biological
AC, Hirschhorn JN, Baron J. Synthesizing genome- cell renal cell carcinoma: biomarker discovery and fluids by anion-exchange chromatography cou-
wide association studies and expression microar- identification of potential controls and conse- pled to ELISA and identification by mass spec-
ray reveals novel genes that act in the human quences of miRNA dysregulation. J Urol 2011; trometry. Proteomics 2012;12:799 – 809.
growth plate to modulate height. Hum Mol Genet 186:1077– 83. 54. Makawita S, Smith C, Batruch I, Zheng Y, Ruckert
2012;21:5193–201. 37. Yousef GM. Personalized cancer genomics: the F, Grutzmann R, et al. Integrated proteomic pro-
21. Warren H, Dudbridge F, Fletcher O, Orr N, John- road map to clinical implementation. Clin Chem filing of cell line conditioned media and pancre-
son N, Hopper JL, et al. 9q31.2-rs865686 as a 2012;58:661–3. atic juice for the identification of pancreatic can-
susceptibility locus for estrogen receptor-positive 38. Mischel PS, Shai R, Shi T, Horvath S, Lu KV, Choe cer biomarkers. Mol Cell Proteomics 2011;10:
breast cancer: evidence from the Breast Cancer G, et al. Identification of molecular subtypes of M111.
Association Consortium. Cancer Epidemiol Bio- glioblastoma by gene expression profiling. Onco- 55. Bauer JA, Chakravarthy AB, Rosenbluth JM, Mi D,
markers Prev 2012;21:1783–91. gene 2003;22:2361–73. Seeley EH, De Matos Granja-Ingram N, et al.
22. Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, 39. Lapuk A, Marr H, Jakkula L, Pedro H, Bhattacha- Identification of markers of taxane sensitivity us-
Haynes C, et al. Complement factor H polymor- rya S, Purdom E, et al. Exon-level microarray ing proteomic and genomic analyses of breast
phism in age-related macular degeneration. Sci- analyses identify alternative splicing programs in tumors from patients receiving neoadjuvant pac-
ence 2005;308:385–9. breast cancer. Mol Cancer Res 2010;8:961–74. litaxel and radiation. Clin Cancer Res 2010;16:
23. Johnson AD, O’Donnell CJ. An open access data- 40. Youssef YM, White NM, Grigull J, Krizova A, 681–90.
base of genome-wide association results. BMC Samy C, Mejia-Guerrero S, et al. Accurate molec- 56. Geiger T, Madden SF, Gallagher WM, Cox J,
Med Genet 2009;10:6. ular classification of kidney cancer subtypes using Mann M. Proteomic portrait of human breast
24. Comprehensive molecular characterization of hu- microRNA signature. Eur Urol 2011;59:721–30. cancer progression identifies novel prognostic
man colon and rectal cancer. Nature 2012;487: 41. Markert EK, Mizuno H, Vazquez A, Levine AJ. markers. Cancer Res 2012;72:2428 –39.
330 –7. Molecular classification of prostate cancer using 57. Hood L, Flores M. A personal view on systems
25. Hudson TJ, Anderson W, Artez A, Barker AD, Bell curated expression signatures. Proc Natl Acad Sci medicine and the emergence of proactive P4
C, Bernabe RR, et al. International network of U S A 2011;108:21276 – 81. medicine: predictive, preventive, personalized
cancer genome projects. Nature 2010;464: 42. Fendler A, Jung M, Stephan C, Honey RJ, Stewart and participatory. N Biotechnol 2012;29:613–24.
993– 8. RJ, Pace KT, et al. miRNAs can predict prostate 58. Virgin HW, Todd JA. Metagenomics and person-
26. Stephens PJ, Tarpey PS, Davies H, Van LP, Green- cancer biochemical relapse and are involved in alized medicine. Cell 2011;147:44 –56.
man C, Wedge DC, et al. The landscape of cancer tumor progression. Int J Oncol 2011;39:1183–92. 59. Girgis AH, Iakovlev VV, Beheshti B, Bayani J,
genes and mutational processes in breast cancer. 43. Heinrich MC, Owzar K, Corless CL, Hollis D, Bor- Squire JA, Bui A, et al. Multilevel whole genome
Nature 2012;486:400 – 4. den EC, Fletcher CD, et al. Correlation of kinase analysis reveals candidate biomarkers in clear
27. Kanehisa M, Goto S, Sato Y, Furumichi M, Tanabe genotype and clinical outcome in the North cell renal cell carcinoma. Cancer Res 2012;72:
M. KEGG for integration and interpretation of American Intergroup Phase III Trial of imatinib 5273– 84.

166 Clinical Chemistry 59:1 (2013)


Genomic Medicine in Cancer
Reviews

60. Weston AD, Hood L. Systems biology, proteomics, other cancers: a review. Clin Chem 2005;51: Leary RJ, et al. The genomic landscapes of human
and the future of health care: toward predictive, 494 –503. breast and colorectal cancers. Science 2007;318:
preventative, and personalized medicine. J Pro- 66. Halling KC, Schrijver I, Persons DL. Test verifi- 1108 –13.
teome Res 2004;3:179 –96. cation and validation for molecular diagnostic 71. Gerlinger M, Rowan AJ, Horswell S, Larkin J,
61. Haber DA, Settleman J. Cancer: drivers and pas- assays. Arch Pathol Lab Med 2012;136: Endesfelder D, Gronroos E, et al. Intratumor het-
sengers. Nature 2007;446:145– 6. 11–3. erogeneity and branched evolution revealed by
62. Amos J, Grody W. Development and integration 67. Sturgeon CM, Hoffman BR, Chan DW, Ch’ng SL, multiregion sequencing. N Engl J Med 2012;366:
of molecular genetic tests into clinical practice: Hammond E, Hayes DF, et al. National Academy 883–92.
the US experience. Expert Rev Mol Diagn 2004; of Clinical Biochemistry Laboratory Medicine 72. Clayton EW. Ethical, legal, and social implications
4:465–77. Practice Guidelines for use of tumor markers in of genomic medicine. N Engl J Med 2003;349:
63. Diamandis EP, Hoffman BR, Sturgeon CM. Na- clinical practice: quality requirements. Clin Chem 562–9.
tional Academy of Clinical Biochemistry Labo- 2008;54:e1–10. 73. Bunnik EM, Schermer MH, Janssens AC. The role
ratory Medicine Practice Guidelines for the Use 68. Kung JT, Gelbart ME. Getting a head start: the of disease characteristics in the ethical debate on
of Tumor Markers. Clin Chem 2008;54:1935–9. importance of personal genetics education in personal genome testing. BMC Med Genomics
64. Ng PC, Murray SS, Levy S, Venter JC. An agenda high schools. Yale J Biol Med 2012;85:87–92. 2012;5:4.
for personalized medicine. Nature 2009;461: 69. Kohane IS, Shendure J. What’s a genome worth? 74. Salari K, Watkins H, Ashley EA. Personalized
724 – 6. Sci Transl Med 2012;4:133fs13. medicine: hope or hype? Eur Heart J 2012;33:
65. Duffy MJ. Predictive markers in breast and 70. Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, 1564 –70.

Clinical Chemistry 59:1 (2013) 167

You might also like