Vous êtes sur la page 1sur 11

Chapter 1

Molecular Diagnostics: Past, Present,


and Future
G.P. Patrinos1, P.B. Danielson2, 3 and W.J. Ansorge4
1
University of Patras School of Health Sciences, Patras, Greece; 2University of Denver, Denver, CO, United States; 3Center for Forensic Science
Research and Education, Willow Grove, PA, United States; 4Ecole Polytechnique Federal Lausanne, EPFL, Lausanne, Switzerland

1.1 INTRODUCTION recurrent episodes of acute pain due to vessel occlusion. In


principle, their findings set the foundations of molecular
Molecular (or nucleic acid-based) diagnosis of human diagnostics, although the big revolution occurred many
disorders is referred to as the detection of genomic variants years later. At that time, when molecular biology was only
that are pathogenic and/or benign in DNA and/or RNA hectically expanding, the provision of molecular diagnostic
samples in order to facilitate detection, diagnosis, sub- services was inconceivable and technically not feasible.
classification, prognosis, and monitoring response to ther- The first seeds of molecular diagnostics were provided in
apy. Molecular diagnostics combines laboratory medicine the early days of recombinant DNA technology, with many
with the knowledge and technology of molecular genetics scientists from various disciplines working in concert.
and has been enormously revolutionized over the last de- cDNA cloning and sequencing were at that time invaluable
cades, benefiting from discoveries in the fields of molecular tools for providing basic knowledge on the primary
biology and genomic technologies (Table 1.1). The iden- sequence of various genes. The latter provided a number of
tification and fine characterization of the genetic basis of DNA probes, allowing for analysis via southern blotting
inherited diseases is vital for the accurate provision of of genomic regions, leading to the concept and application
diagnosis. Gene discovery, via high-throughput methods, of restriction fragment length polymorphism (RFLP) to
such as next-generation sequencing or genome-wide asso- track a variant allele from heterozygous parents to a high-
ciation studies, provides invaluable insights into the risk pregnancy. In 1976, Kan and coworkers carried out,
mechanisms of disease, and genomic markers allow phy- for the first time, prenatal diagnosis of a-thalassemia, using
sicians to not only assess disease predisposition but also to hybridization on DNA isolated from fetal fibroblasts. Also,
design and implement accurate diagnostic methods. The Kan and Dozy (1978), implemented RFLP analysis to
latter is of great importance, as the plethora and variety of pinpoint sickle cell alleles of African descent. This break-
molecular defects demands the use of multiple rather than a through provided the means of establishing similar diag-
single variant detection platform. Molecular diagnostics has nostic approaches for the characterization of other genetic
gradually become a clinical reality with its roots deep into diseases, such as phenylketonurea (Woo et al., 1983), cystic
the basic science of gene expression and gene function. fibrosis (Farrall et al., 1986), and so on.
At that time, however, a significant technical bottleneck
1.2 HISTORY OF MOLECULAR had to be overcome. The identification of the pathogenic
variant was possible only through the construction of a
DIAGNOSTICS: INVENTING THE WHEEL
genomic DNA library from the affected individual, in order
In 1949, Pauling and his coworkers introduced the term to first clone the variant allele and then determine its
molecular disease in the medical vocabulary, based on their nucleotide sequence. Again, many human globin gene
discovery that a single amino acid change at the b-globin mutations were among the first to be identified through
chain leads to sickle cell anemia, characterized mainly by such approaches (Busslinger et al., 1981; Treisman et al.,

Molecular Diagnostics. http://dx.doi.org/10.1016/B978-0-12-802971-8.00001-8 1


Copyright © 2017 Elsevier Ltd. All rights reserved.
2 Molecular Diagnostics

TABLE 1.1 Timeline of the Principal Discoveries in the Field of Molecular Biology, Which Influenced the
Development of Molecular Diagnostics
Date Discovery
1949 Characterization of sickle cell anemia as a molecular disease
1953 Discovery of the DNA double helix
1958 Isolation of DNA polymerases
1960 First hybridization techniques
1969 In situ hybridization
1970 Discovery of restriction enzymes and reverse transcriptase
1975 Southern blotting
1977 DNA sequencing
1983 First synthesis of oligonucleotides
1985 Restriction fragment length polymorphism analysis
1985 Invention of the polymerase chain reaction
1986 Development of fluorescent in situ hybridization
1988 Discovery of the thermostable DNA polymerasedoptimization of the polymerase chain reaction
1992 Conception of the real-time polymerase chain reaction
1993 Discovery of structure-specific endonucleases for cleavage assays
1996 First application of DNA microarrays
2001 First draft versions of the human genome sequence
2001 Application of protein profiling in human diseases
2002 Launch of the HapMap project
2005 Introduction of high-throughput next-generation sequencing technology
2008 Launch of the 1000 Genomes Project
2013 Introduction of the CRISPR system for gene editing
2014 Announcement of the sequencing of the human genome for $1000
2015 Launch of the Precision Medicine Initiative by US President Barack Obama

1983). In 1982, Orkin and his coworkers showed that a developed, setting the basis for variant screening and
number of sequence variations were linked to specific scanning methods. The first methods involved mismatch
pathogenic HBB gene variants. These groups of RFLPs, detection in DNA/DNA or RNA/DNA heteroduplexes
termed haplotypes (both intergenic and intragenic), have (Myers et al., 1985a,b) or differentiation of mismatched
provided a first-screening approach in order to detect a DNA heteroduplexes using gel electrophoresis, according
disease-causing variant. Although this approach enabled to their melting profile (Myers et al., 1987). Using this
researchers to predict which HBB allele was pathogenic, laborious and time-consuming approach, a number of
significantly facilitating mutation screening, no one was in variant sequence alleles have been identified, which made
the position to determine the exact nature of the disease- possible the design of short synthetic oligonucleotides that
causing mutation, as many different HBB gene variants were used as allele-specific probes onto genomic Southern
were linked to a specific haplotype in different populations blots. This experimental design was quickly implemented
(further information is available at http://globin.bx.psu.edu/ for the detection of b-thalassemia mutations (Orkin et al.,
hbvar; Patrinos et al., 2004; Giardine et al., 2014). 1983; Pirastu et al., 1983).
At the same time, in order to provide a shortcut to DNA Despite intense efforts from different laboratories
sequencing, a number of exploratory methods for pin- worldwide, the diagnosis of inherited diseases on the DNA
pointing pathogenic variants in patients’ DNA were level was still underdeveloped and therefore still not ready
Molecular Diagnostics: Past, Present, and Future Chapter | 1 3

to be implemented in clinical laboratories for routine activity of resolvase enzymes T4 endonuclease VII
analysis of patients due to the complexities, costs, and time and T7 endonuclease I to digest heteroduplex DNA
requirements of the technology available. It was only after a formed by annealing wild type and mutant DNA
few years that molecular diagnosis entered its golden era (Mashal et al., 1995). Digestion fragments indicate
with the discovery of the most powerful molecular biology the presence and the position of any variants. A vari-
tool since cloning and sequencing, the polymerase chain ation of the theme involves the use of chemical agents
reaction (PCR). for the same purpose (Saleeba et al., 1992). Another
enzymatic approach for variant detection is the oligo-
1.3 THE POST-POLYMERASE CHAIN nucleotide ligation assay (Landegren et al., 1988;
Chapter 3); ligation was also one of the main princi-
REACTION REVOLUTION ples of one of the most widely used next-generation
The discovery of PCR (Saiki et al., 1985; Mullis and sequencing approaches (see also Chapter 8).
Faloona, 1987) and its quick optimization, using a ther- 2. Electrophoretic-based techniques. This category is
mostable Taq DNA polymerase from Thermus aquaticus characterized by a plethora of different approaches
(Saiki et al., 1988), has greatly facilitated and in principle designed for the screening of known or unknown muta-
revolutionized molecular diagnostics. The most powerful tions, based on the different electrophoretic mobility of
feature of PCR is the large amount of copies of the target the mutant alleles, under denaturing or nondenaturing
sequence generated by its exponential amplification, which conditions. Single-strand conformation polymorphism
allows the identification of a known mutation within a (SSCP) and heteroduplex analyses (HDA; Orita et al.,
single day, rather than months. Also, PCR has markedly 1989; see Chapter 3) were among the first methods
decreased or even diminished the use of radioactivity for designed to detect molecular defects in genomic loci.
routine molecular diagnosis. This has allowed molecular In combination with capillary electrophoresis, SSCP
diagnostics to enter the clinical laboratory for the provision and HDA analysis now provide an excellent, simple,
of genetic services, such as carrier or population genetic and rapid variant detection platform with low operation
screening, prenatal diagnosis of inherited diseases, or, in costs and, most interestingly, the potential of easily be-
recent years, the identification of unknown variants, in ing automated, thus allowing for high-throughput anal-
close collaboration with research laboratories. Therefore by ysis of patients’ DNA. Similarly, denaturing gradient
being moved to their proper environment, the clinical lab- gel electrophoresis (DGGE) and temperature gradient
oratory, molecular diagnostics could provide the services gel electrophoresis can be used equally well for variant
for which they have been initially conceived. allele detection (see Chapter 3). In this case, electropho-
The discovery of PCR also has provided the founda- retic mobility differences between a wild type and
tions for the design and development of many variant variant allele can be “visualized” in a gradient of dena-
detection schemes, based on amplified DNA. In general, turing agents, such as urea and formamide, or of
PCR is either used for the generation of DNA fragments to increasing temperature. A less common variant detec-
be analyzed or is part of the detection method. The first tion technique is two-dimensional gene scanning, based
attempt was the use of restriction enzymes (Saiki et al., on two-dimensional electrophoretic separation of ampli-
1985) or oligonucleotide probes, immobilized onto mem- fied DNA fragments, according to their size and base
branes or in solution (Saiki et al., 1986), in order to detect pair sequence. The latter involves DGGE, following
the existing genetic variation, in particular the sickle cell the size separation step.
disease-causing mutation. In the following years, an even 3. Solid phase-based techniques. This set of techniques
larger number of variant detection approaches have been consists of the basis for most of the present-day muta-
developed and implemented (see also Chapter 3). These tion detection technologies, since they have the extra
techniques can be divided roughly into three categories, advantage of being easily automated and hence are
depending on the basis for discriminating the allelic highly recommended for high-throughput mutation
variants: detection or screening. A fast, accurate, and convenient
method for the detection of known mutations is reverse
1. Enzymatic-based methods. RFLP analysis was histori-
dot-blot, initially developed by Saiki et al. (1989) and
cally the first widely used approach, exploiting the al-
implemented for the detection of HBB gene variants
terations in restriction enzyme sites, leading to the
leading to b-thalassemia. The essence of this method
gain or loss of restriction events (Saiki et al., 1985). is the utilization of oligonucleotides, bound to a mem-
Subsequently, a number of enzymatic approaches for
brane, as hybridization targets for amplified DNA.
variant allele detection have been conceived, based
Some of this technique’s advantages are that one mem-
on the dependence of a secondary structure on the pri-
brane strip can be used to detect many different known
mary DNA sequence. These methods exploit the
mutations in a single individual (a one strip-one patient
4 Molecular Diagnostics

type of assay), the potential of automation, and the ease disciplines to intensify their research efforts to improve by
of interpretation of the results, using a classical avidin- orders of magnitude the existing methods for genomic
biotin system. However, this technique cannot be used variant detection, to make available data sets with genomic
for the detection of unknown mutations. Continuous variation and analyze these sets using specialized software,
development has given rise to allele-specific hybridiza- to standardize and commercialize genetic tests for routine
tion of amplified DNA [PCR-ASO (Allele Specific diagnosis, and to improve the existing technology in order
Oligonucleotide), Chapter 3] on filters, recently to provide state-of-the-art automated devices for high-
extended to DNA oligonucleotide microarrays (see throughput genetic analysis.
Chapter 18) for high-throughput mutation analysis The biggest challenge, following the publication of the
(Gemignani et al., 2002; Cremonesi et al., 2007). In human genome draft sequence, was to improve the existing
particular, oligonucleotides of known sequence are variant detection technologies to achieve robust, cost-
immobilized onto appropriate surfaces, and hybridiza- effective, rapid, and high-throughput analysis of genomic
tion of the targets to the microarray is detected, mostly variation. Also, the increased pace of novel variant detec-
using fluorescent dyes. tion and gene discovery dictated the harmonization of gene
nomenclature, an effort that was spearheaded by the Human
The choice of the variant detection method is dependent
Genome Variation Society (http://www.hgvs.org; see also
upon a number of variables, including the variation spec-
Chapter 2). Since 2005, genomic technology has improved
trum of a given inherited disorder, the available infra-
rapidly, and new high-throughput variant detection tech-
structure, the number of tests performed in the diagnostic
niques have become available, whereas old methodologies
laboratory, and issues of intellectual properties (see also
have evolved to fit into the increasing demand for auto-
Section 1.5.1). Most of the clinical diagnostic laboratories
mated and high-throughput screening or were gradually
have not invested in expensive high-technology in-
abandoned. The chromatographic detection of polymorphic
frastructures, since the test volumes (the number of tests)
changes of pathogenic variants using denaturing high-
expected to be performed have not been large enough to
performance liquid chromatography (DHPLC; for review,
justify the capital investment. Therefore simple “home-
see Xiao and Oefner, 2001) is one of the new technologies
brew”screening tests such as SSCP and HDA were and
that emerged. DHPLC reveals the presence of a genetic
still are the methods of choice for many clinical labora-
variation by the differential retention of homo- and het-
tories, as they allow for rapid and simultaneous detection of
eroduplex DNA on reversed phase chromatography under
different sequence variations at a detection rate of close to
partial denaturation. Single-base substitutions, deletions,
100%. Although DNA amplification has significantly
and insertions can be detected successfully by ultraviolet or
facilitated the expansion of molecular diagnostics, it
fluorescence monitoring within 2 to 3 min in unpurified
nonetheless has a number of limitations, such as amplifi-
PCR products as large as 1.5-kilo bases. These features,
cation of cytidine-guanine repeat-rich regions, the error-
together with its low cost, make DHPLC one of the most
prone features of Taq polymerase (at a range of 104 to
powerful tools for mutational analysis. Also, pyrose-
105 per nucleotide), and so on. Finally, it is noteworthy
quencing, a nongel-based genotyping technology, provides
that despite the wealth of variant detection methodologies,
a very reliable method and an attractive alternative to
DNA sequencing, particularly in the era of whole genome
DHPLC. Pyrosequencing detects de novo incorporation of
sequencing and the breakthroughs in next-generation
nucleotides based on the specific template. The incorpora-
sequencing technology, is considered the golden standard
tion process releases a pyrophosphate, which is converted
and the definitive experimental procedure for variant call-
to ATP and followed by luciferase stimulation. The light
ing. However, the costs for the initial investment and the
produced, detected by a charge couple device camera, is
difficulties for standardization and interpretation of
“translated” to a pyrogram, from which the nucleotide
ambiguous results have restricted its use especially to basic
sequence can be deducted (Ronaghi et al., 1998). This
research laboratories.
approach constituted the basis for the development of the
1.4 MOLECULAR DIAGNOSTICS IN THE first next-generation sequencing approaches by 454 Life
Technologies (see also Chapter 8).
POST-GENOMIC ERA
One of the major advances was the invention of the
In February 2001, with the announcement of the first draft real-time PCR and the numerous variations of this theme
sequence of the human genome (International Human (Holland et al., 1991; see Chapter 4). The method allows
Genome Sequencing Consortium, 2001; Venter et al., for the direct detection of the PCR product during the
2001) and subsequently with the genomic sequence of exponential (mid-log) phase of the reaction, therefore
other organisms, molecular biology has entered into a new combining amplification and detection in one single step.
era with unprecedented opportunities and challenges. These The increased speed of real-time PCR is due largely to
tremendous developments put pressure on a variety of reduced cycles, the removal of post-PCR detection
Molecular Diagnostics: Past, Present, and Future Chapter | 1 5

procedures, and the use of fluorogenic labels and sensi- carcinomas of the oral cavity (Knezevic et al., 2001) or the
tive methods of detecting their emissions. Therefore real- identification of proteins that induce an acute antibody
time PCR is a very accurate and sensitive methodology response in autoimmune disorders, using auto-antigen
with a variety of applications in molecular diagnostics, arrays (Robinson et al., 2002), emerged in the early
allows a high-throughput, and can easily be automated 2000s, indicating that proteomic pattern analysis ulti-
and performed on very small volumes, which makes it mately might be applied as a screening tool for cancer in
the method of choice for many modern diagnostic high-risk and general populations.
laboratories. The development of state-of-the-art variant detection
Above all, the DNA microarray-based genotyping platforms not only has a positive impact on molecular ge-
approaches offer simultaneous analysis of many sequence netic testing of inherited disorders but also provides tech-
alterations (see Chapter 18). In particular, microarrays nical means to other disciplines, such as to ascertain
consist of hundreds of thousands up to millions of oli- genetically modified (GM) products, which may contami-
gonucleotides attached on a solid surface in an ordered nate non-GM seeds, or food ingredients containing addi-
array fashion. The DNA sample of interest is PCR- tives and flavorings that have been genetically modified or
amplified and then hybridized onto the microarray. Each have been produced from GM organisms, or the genotype
oligonucleotide in the high-density array acts as an allele- of an animal strain. Molecular genetic testing is also
specific probe, and therefore perfectly matched sequences applicable in the individualization of drug doses, also
hybridize more efficiently to their corresponding oligo- known as pharmacogenomics (Chapter 16), referred to as
nucleotides on the array. The hybridization signals are the delineation of interindividual genetic variability in
quantified by high-resolution fluorescent scanning and genes that are mainly involved in drug metabolism and
analyzed by computer software, resulting in the identifi- transport with drug efficacy and adverse effects. This
cation of the genotype in the corresponding places in the approach amalgamates technological expertise from high-
genome. Therefore using a high-density microarray makes throughput omics approaches, such as genomics, tran-
possible the simultaneous detection of a great number of scriptomics, and functional genomics, to define and predict
DNA alterations, hence facilitating genome-wide the nature of the response of an individual to a drug
screening. treatment and to rationally design newer drugs or improve
There has been a significant development of prote- existing ones (Squassina et al., 2010). The same can be also
omics, which has the potential to become an indispensable applicable in the personalization of diet, in an emerging
tool for molecular diagnostics. A useful repertoire of discipline known as Nutrigenomics (Chapter 17). Ulti-
proteomic technologies is available, with the potential to mately, the identified genomic sequence variants need to be
undergo significant technological improvements, which organized and stored in well-curated and specialized variant
would be beneficial for increased sensitivity and databases, enabling a physician or researcher to query and
throughput while reducing the sample requirement (see retrieve information relevant to diagnostic issues (see
Chapter 13). The improvement of these technologies is a Chapter 20).
significant advance toward the need for better disease di- Finally, DNA analysis and testing have also signifi-
agnostics. The detection of disease-specific protein pro- cantly revolutionized the forensic sciences. Technical
files goes back to the use of two-dimensional protein gels advances in molecular biology and increasing knowledge
(Hanash, 2000), when it was demonstrated that leukemias of the human genome have had a major impact on
could be classified into different subtypes based on the forensic medicine (see Chapter 21). Genetic character-
different protein profile (Hanash et al., 2002). Nowadays, ization of individuals at the DNA level enables identity
mass spectrometers are able to resolve many protein and testing from a minimal amount of biological specimen,
peptide species in body fluids, being virtually set to such as hair, blood, semen, bone, and so forth, in cases of
revolutionize protein-based disease diagnostics (see sexual assault, homicide, and unknown human remains,
Chapter 13). The robust and high-throughput nature of the and paternity testing is also changing from the level of
mass spectrometric instrumentation is unparalleled and gene products to the genomic level. DNA testing is by far
imminently suited for future clinical applications, as more advantageous over conventional forensic methods
elegantly demonstrated by many retrospective studies in and over the years has contributed to the acquittal of
cancer patients (reviewed in Petricoin et al., 2002). Also, falsely accused people (saving most of them even from
high-throughput protein microarrays, constructed from death row), the identification of individuals who
recombinant, purified, and yet functional proteins, allow committed criminal acts (Cohen, 1995), and even helped
the miniaturized and parallel analysis of large numbers of to specify identities of unknown human remains, such as
diagnostic markers in complex samples. The first pilot those from the victims at Ground Zero in New York or
studies on disease tissues, such as assessing protein from the skeletons of the Romanov family members (Gill
expression profiles in tissue derived from squamous cell et al., 1994; see Chapter 22).
6 Molecular Diagnostics

1.5 FUTURE PERSPECTIVES: WHAT LIES plethora of variant detection methods is available, “. the
BEYOND best platform is the one that works best in your own
laboratory.” There is ample choice for genotyping, such as
Molecular diagnostics stem back to the spring of 1953, filters, gels, microarrays, and microtiter plates, for
when the DNA double helix was first announced. Today, amplification-based technologies, for separation tech-
molecular diagnostics embody a set of high-throughput niques, such as blotting, capillary electrophoresis, micro-
technological advances, which reveal the genotype in arrays, and mass spectroscopy, and finally different means
thousands of genomic positions and the entire genome at a for labeling, such as radioactive, fluorescent, chemilumi-
very high accuracy and gradually decreasing costs. This nescent, or enzymatic substances. The variety of detection
also leads to a better understanding of the basis of inherited approaches makes it difficult to determine which one is
diseases, therefore allowing molecular diagnostics to play a better suited for a laboratory setting. Generally speaking,
key role in patient or disease management. Presently, a DNA sequencing, especially in the next-generation
great number of samples are analyzed annually worldwide sequencing era, is the golden standard for the identifica-
in both public and private laboratories, and the number of tion of, causative or benign, DNA sequence variations (see
genetic tests available is steadily increased year by year, also Chapter 9), but then again different issues have to be
making molecular diagnostic laboratories indispensable in addressed such as the sequencing coverage, the enrichment
laboratory medicine. With the existing sequencing plat- methods, the genes to be analyzed, and so forth. The initial
forms, newborns can be screened for a number of inherited investment costs and the expected test volume are some of
treatable diseases, while it is also possible that in the not- the factors that need to be taken into consideration prior to
so-distant future, children at high risk for coronary artery choosing the detection technique. Related issues are the
disease will be identified and treated to prevent changes in costs of the adjacent hardware and software, testing re-
their vascular walls during adulthood. Similarly, parents agents, and kits. The latter is of great importance, since
will have the option of being informed about their carrier most of the diagnostic laboratories today are running
status for many recessive diseases before they decide to “home-brew” assaysdfor example, not using well-
start a family. Also, for middle-aged and older populations, standardized genetic testing kits due to cost barriers,
scientists will be able to determine risk profiles for various which brings to surface the issue of quality control of the
late-onset diseases, preferably before the appearance of reagents (see Chapter 29) and safety (see Chapter 28).
symptoms, which at least could be partly prevented through Another very important issue is training the personnel
dietary (Chapter 17) or pharmaceutical interventions of a molecular diagnostic laboratory, reflected in the quality
(Chapter 16). In the near future, the preemptive genotyping and the correct interpretation of results. Continuous genetic
of genes involved in drug metabolism and transport will education of the personnel of the diagnostic laboratory is
help toward individualizing drug response and will become crucial for the accuracy of the results provided, particularly
indispensable in standard medical practice. Although the in recently emerged disciplines such as Pharmacogenomics
majority of these issues are gradually becoming a reality, (Reydon et al., 2012; Kampourakis et al., 2014). Many
some of them are still based on promises, though quite times, such as in the case of prenatal or preimplantation
optimistic ones. Thus some of the new perspectives of the diagnosis, irrevocable decisions need to be made, most of
field could simply be a decade away. the time based on a simple test result. As a result of
continuous training and proficiency testing schemes, there
1.5.1 Commercializing Molecular has been a significant reduction of the number of incorrect
Diagnostics genotypes diagnosed (http://www.eurogentest.org). In the
United States, there is a voluntary biannual proficiency test
Currently, clinical molecular genetics is part of mainstream for molecular diagnostic laboratories, while in Europe, the
health care worldwide with a molecular diagnostic unit or EuroGenTest European Network of Excellence (http://
department within each health care unit. Although the www.eurogentest.org) has been founded to promote qual-
notion of molecular diagnostics has increasingly gained ity in molecular genetic testing through the provision of
momentum, genetic tests are still not generally used for external quality assessment (proficiency testing schemes)
population screening but rather for diagnosis, carrier and the organization of best practice meetings and publi-
screening, and prenatal diagnosis and only on a limited cation of guidelines. It is generally true that many geneti-
basis. The lack of cost-effectiveness analyses is one of the cists and nongeneticist physicians would benefit from
reasons (Snyder et al., 2014). Therefore in order to make continuous genetics education regarding the appropriate
molecular diagnostics widely available, several obstacles use of molecular diagnostic tests, which is necessary to
and issues need to be taken into consideration and resolved. evaluate the method preanalytically and to interpret results.
The first important issue is the choice of the technology Legal considerations and ethical concerns are also
and variant detection platform. Despite the fact that a hurdles that need to be overcome. One issue is the
Molecular Diagnostics: Past, Present, and Future Chapter | 1 7

reimbursement of diagnosis costs. At present, very few tests based on their genetic background. Some of the reasons for
are reimbursed by insurance companies, and the necessary many types of adverse drug reactions are already known
regulatory and legal framework is far from complete. and are often related to polymorphic gene alleles of drug-
“Legalizing” molecular testing by the adoption of relevant metabolizing enzymes (Squassina et al., 2010). The
regulations would probably result in an increase in test application of high-throughput genotyping tools for the
volumes, and at the same time it can pose an immense identification and screening of single nucleotide poly-
barrier to uncontrolled genetic testing. Similarly, the need morphisms can eventually lead to the determination of the
to obtain informed consent from the patient to be analyzed unique molecular signature of an individual in a relatively
is also of great importance and should be encouraged and short period of time, especially with the advent of whole
facilitated by the diagnostic laboratory, particularly in the genome sequencing and its application in pharmacoge-
case of incidental findings that arise from next-generation nomics (Mizzi et al., 2014; Katsila and Patrinos, 2015).
sequencing approaches (Roche and Berg, 2015). This way, individual drug responses can be predicted
On the other hand, the issue of intellectual properties from predetermined genetic variances correlated with a
hampers the wide commercialization of molecular di- drug effect. In other words, this will allow the physician to
agnostics. Almost all the clinically relevant genes have provide the patient with a selective drug prescription (see
been patented, and the terms that the patent holders offer Chapter 16). Also, specialized information technology
vary considerably. Among the difficulties are the limiting systems will allow for translation of this information into a
choice of variant detection platforms, the large loyalties for clinically meaningful format, which clinicians will then use
reagent use, and the exclusive sublicenses that many to evaluate and adjust the drug dose to each patient, hence
companies grant to clinical laboratories, leading to mo- allowing for a complete patient analysis and drug evalua-
nopolies. Since one of the biggest challenges that the tion, especially in a preemptive manner (Lakiotaki et al.,
clinical laboratory is facing is patent and regulatory 2016). In addition to these efforts, there is a growing need
compliance, partnerships and collaborations may be to incorporate this increasingly complex body of
envisaged in order to take technology licenses to the knowledge to standard medical practice. Incorporating
diagnostic laboratory that will subsequently develop, stan- pharmacogenomics-related courses into the standard cur-
dardize, and distribute the assays. This will partly alleviate riculum of medical schools can potentially ensure that the
some of the intellectual properties issues. Finally, the issue forthcoming generation of clinicians and researchers will be
of the medical genetics specialty is more urgent than ever. familiar with the latest developments in that field and will
In the United States, medical genetics has been formally be capable of providing patients with the expected benefits
recognized as a medical specialty only within the past of personalized medicine (Pisanu et al., 2014).
15 years, and in Europe, medical genetics only recently has Similarly, nutrigenomics (or nutritional genomics) in-
been formally recognized as a specialty (http://www.eshg. vestigates the interactions between nutrition and an in-
org). The implementation of this decision is still facing dividual’s genome and the consequent downstream effects
substantial difficulties (http://www.eshg.org/ on their phenotype with the aim to provide tailored nutri-
geneticseurope.htm), which will probably take years to tional advice or develop specialist food products (see
bypass. With the completion of the Human Genome Proj- Chapter 17). In other words, nutrigenomics recognizes that
ect, genetics has become the driving force in medical a specific dietary advice that can be beneficial for one in-
research and is now poised for integration into medical dividual may be inappropriate, or actually harmful, to
practice. An increase in the medical genetics workforce, another. Although comparable to pharmacogenomics,
including geneticists and genetic counselors, will be nutrigenomics is still considered to be an emerging science,
necessary, while the same is true with the emergence of contrary to pharmacogenomics that is considered to have
new disciplines, such as genome informatics, needed for “come of age” (Pavlidis et al., 2015).
genome results interpretation (Potamias et al., 2014; see However, there are growing concerns on the ethical
also Chapter 19). After all, the Human Genome Project has aspects of personalized medicine. First of all, equality in
made information of inestimable diagnostic and therapeutic medical care needs to be ensured, when genetics foretell
importance available, and the medical profession now has clinicians which patients would be less likely to benefit
the obligation to rise to both the opportunities and chal- from a particular drug treatment. Second, it will become
lenges that this wealth of genetic information presents. increasingly vital to devise operational tools for the pre-
vention of stigmatization and discrimination of different
populations, in particular on ethnic grounds (van Ommen,
1.5.2 Personalized Medicine
2002), and therefore every precaution should be taken to
The term “personalized medicine” refers to the practice of eliminate all lingering prejudice and bias associated with
medicine where patients receive the most appropriate the study of human genetic variation. Other dilemmas
medical treatment, fitting dosage, and combination of drugs include the right to deny an available treatment to specific
8 Molecular Diagnostics

patient populations according to genetic-derived in- metabolomics, transcriptomics, and metagenomics, medi-
dications, as is currently the case with prenatal diagnosis. ated by accurate genome informatics translation services
(Fig. 1.1).
Even though the expectations are high and companies
1.5.3 Personal Genomics
are using these new technologies to provide information to
The ultimate goal in health care will be the efficient inte- individuals to predict health and disease outcomes, even
gration of molecular diagnostics with therapeutics. With the behavioral traits, it is generally premature to make promises
advent of next-generation sequencing in 2005 (Margulies for clinically useful information from genomic analyses.
et al., 2005) and the avalanche of developments in this field Next to that, there is an inherent danger of overestimating
since then (see Chapters 8 and 9), experts believe that the usefulness of the various personalized genomic tests,
reasonably soon, people will be able to have their own particularly those that can be ordered directly by con-
genomes sequenced for under $1000. As a matter of fact, sumers, also known as direct-to-consumer (DTC) genetic
there are already claims that the human genome can be tests (Magnus et al., 2009). Unlike other genetic analyses,
sequenced for $1000 with a low depth of coverage. This is these tests provide a sheer amount of genetic information,
going to involve sequencing technology that is much but their diagnostic or prognostic value remains uncertain
cheaper and faster than today’s machines, both commer- because of the lack of information about the influence of
cially available and prototypes, and several efforts are environmental and other factors and the weak association
currently under way, often encouraged by major funding. for the vast majority of genetic loci with disease. Several
Previously, the entire DNA sequence of only a handful of studies and metaanalyses suggest that there is insufficient
individuals had been sequenced, such as Craig Venter scientific evidence to conclude that genomic profiles are
(Levy et al., 2007), Jim Watson (Wheeler et al., 2008), and useful in measuring the genetic risk for common diseases or
so on, while today there are thousands of genomes that in developing personalized diet (Pavlidis et al., 2015; 2016)
have been sequenced, providing useful insights into the and lifestyle recommendations for disease prevention
genetic basis of human diseases. Yet, several million ge- (Patrinos et al., 2013). It is puzzling how some companies
nomes should be sequenced so that further insights can be in the DTC sector use their clients’ genetic profiles to tailor
gained on how to better individualize treatment modalities individualized diets and lifestyle recommendations. Also, it
(Table 1.2). In the future, a person may appear at the clinic is noteworthy that some of the companies that provide
for treatment, “carrying” his or her entire genome at hand, personal genomic tests have no physicians involved in
or alternatively, nanotechnology could eventually enable ordering these tests, with the argument that “.patients
DNA analysis with a portable DNA sequencing device. deserve direct access to their health information without a
Similarly, a personal genomics health coach may be able to physician intermediary” (http://www.nytimes.com/2008/
provide timely information regarding one’s health, based 06/26/business/26gene.html). In a few cases, these com-
on both artificial intelligence- and machine learning-aided panies were banned by regulatory authorities from further
algorithms and the wealth of genomics data available in selling their services. Overall, there are questions as to
the literature (see also Chapter 19). Overall, the provision whether and how to regulate these tests, about the extent to
of accurate personalized medicine services will comprise a which they provide (useful) medical information, and the
complex interplay between (pharmaco)genomics, risks from misinterpreting them.

TABLE 1.2 Impact of Human Genome Sequencing in Advancing Knowledge in Human Genetics and Personalized
Medicine
Number of
genomes Impact
Thousands l Identification of human genomic variations
l Technology development for the complete and error-free sequencing of the human genome for patients
and the general population
Millions l Understanding of the molecular basis of human genetic diseases
l Optimization of the genotypeephenotype correlation
l Development of software tools for genotypeephenotype correlation in patients’ genomic profiles
Billions l Predictive and personalized medicine
l Treatment individualization
l Improvement of quality of life
l Prevention of human diseases
Molecular Diagnostics: Past, Present, and Future Chapter | 1 9

Farrall, M., Rodeck, C.H., Stanier, P., Lissens, W., Watson, E., Law, H.Y.,
Warren, R., Super, M., Scambler, P., Wainwright, B., Williamson, R.,
1986. First-trimester prenatal diagnosis of cystic fibrosis with linked
DNA probes. Lancet 327, 1402e1405.
Gemignani, F., Perra, C., Landi, S., Canzian, F., Kurg, A., Tonisson, N.,
Galanello, R., Cao, A., Metspalu, A., Romeo, G., 2002. Reliable
detection of beta-thalassemia and G6PD mutations by a DNA
microarray. Clin. Chem. 2002 (48), 2051e2054.
Giardine, B., Borg, J., Viennas, E., Pavlidis, C., Moradkhani, K., Joly, P.,
Bartsakoulia, M., Riemer, C., Miller, W., Tzimas, G., Wajcman, H.,
Hardison, R.C., Patrinos, G.P., 2014. Updates of the HbVar database
of human hemoglobin variants and thalassemia mutations. Nucleic
Acids Res. 42, D1063eD1069.
Gill, P., Ivanov, P.L., Kimpton, C., Piercy, R., Benson, N., Tully, G.,
Evett, I., Hagelberg, E., Sullivan, K., 1994. Identification of the re-
mains of the Romanov family by DNA analysis. Nat. Genet. 6,
130e135.
Hanash, S.M., 2000. Biomedical applications of two-dimensional elec-
trophoresis using immobilized pH gradients: current status. Electro-
FIGURE 1.1 Schematic drawing depicting the interplay among the
various omics disciplines that contribute to the implementation of phoresis 21, 1202e1209.
personalized medicine. Hanash, S.M., Madoz-Gurpide, J., Misek, D.E., 2002. Identification of
novel targets for cancer therapy using expression proteomics. Leu-
kemia 16, 478e485.
1.6 CONCLUSIONS Holland, P.M., Abramson, R.D., Watson, R., Gelfand, D.H., 1991.
Detection of specific polymerase chain reaction product by utilizing
In the coming years, molecular diagnostics will constitute an the 50 e30 exonuclease activity of Thermus aquaticus. Proc. Natl.
integral part of medical practice and public health world- Acad. Sci. U.S.A. 88, 7276e7280.
wide. Molecular genetic testing will facilitate the detection International Human Genome Sequencing Consortium, 2001. Initial
and characterization of human disease as well as monitoring sequencing and analysis of the human genome. Nature 409, 860e921.
the drug response and will assist in the identification of Kampourakis, K., Vayena, E., Mitropoulou, C., Borg, J., van Schaik, R.H.,
genetic modifiers and disease susceptibility. Whole genome Cooper, D.N., Patrinos, G.P., 2014. Key challenges for next genera-
sequencing is likely to become the gold standard in assessing tion pharmacogenomics. EMBO Rep. 15, 472e476.
DNA variation and changes in gene expression. However, Kan, Y.W., Dozy, A.M., 1978. Polymorphism of DNA sequence adjacent
to human beta-globin structural gene: relationship to sickle mutation.
there are major hurdles to overcome before the imple-
Proc. Natl. Acad. Sci. U.S.A. 75, 5631e5635.
mentation of these tests in clinical laboratories, such as
Kan, Y.W., Golbus, M.S., Dozy, A.M., 1976. Prenatal diagnosis of alpha-
which test to employ, the choice of technology and equip- thalassemia. Clinical application of molecular hybridization. N. Engl.
ment, and issues such as cost-effectiveness, accuracy, J. Med. 295, 1165e1167.
reproducibility, personnel training, reimbursement by third- Katsila, T., Patrinos, G.P., 2015. Whole genome sequencing in pharma-
party payers, and intellectual property. Together with cogenomics. Front. Pharmacol. 6, 61.
proteomic-based testing, these advances will improve mo- Knezevic, V., Leethanakul, C., Bichsel, V.E., Worth, J.M., Prabhu, V.V.,
lecular diagnostics and will present additional challenges for Gutkind, J.S., Liotta, L.A., Munson, P.J., Petricoin 3rd, E.F.,
implementing such technology in public or private research Krizman, D.B., 2001. Proteomic profiling of the cancer microenvi-
units, hospitals, clinics, and pharmaceutical companies. ronment by antibody arrays. Proteomics 1, 1271e1278.
Lakiotaki, K., Kartsaki, E., Kanterakis, A., Katsila, T., Patrinos, G.P.,
Potamias, G., 2016. ePGA: a web-based information system for
translational pharmacogenomics. PLoS One. http://dx.doi.org/
REFERENCES
10.1371/journal.pone.0162801 (in press).
Busslinger, M., Moschonas, N., Flavell, R.A., 1981. Beta þ thalassemia: Landegren, U., Kaiser, R., Sanders, J., Hood, L., 1988. A ligase-mediated
aberrant splicing results from a single point mutation in an intron. Cell gene detection technique. Science 241, 1077e1080.
27, 289e298. Levy, S., Sutton, G., Ng, P.C., Feuk, L., Halpern, A.L., Walenz, B.P.,
Cohen, J., 1995. Genes and behavior make an appearance in the O. J. Trial. Axelrod, N., Huang, J., Kirkness, E.F., Denisov, G., Lin, Y.,
Sci. 268, 22e23. MacDonald, J.R., Pang, A.W., Shago, M., Stockwell, T.B.,
Cremonesi, L., Ferrari, M., Giordano, P., Harteveld, C.L., Kleanthous, M., Tsiamouri, A., Bafna, V., Bansal, V., Kravitz, S.A., Busam, D.A.,
Papasavva, T., Patrinos, G.P., Traeger-Synodinos, J., 2007. An Beeson, K.Y., McIntosh, T.C., Remington, K.A., Abril, J.F., Gill, J.,
overview of current microarray-based human globin gene mutation Borman, J., Rogers, Y.H., Frazier, M.E., Scherer, S.W.,
detection methods. Hemoglobin 31, 289e311.
10 Molecular Diagnostics

Strausberg, R.L., Venter, J.C., 2007. The diploid genome sequence of Pavlidis, C., Lanara, Z., Balasopoulou, A., Nebel, J.C., Katsila, T.,
an individual human. PLoS Biol. 5, e254. Patrinos, G.P., 2015a. Meta-analysis of nutrigenomic biomarkers de-
Magnus, D., Cho, M.K., Cook-Deegan, R., 2009. Direct-to-consumer notes lack of association with dietary intake and nutrient-related pa-
genetic tests: beyond medical regulation? Genome Med. 1, 17. thologies. OMICS 19, 512e520.
Margulies, M., Egholm, M., Altman, W.E., Attiya, S., Bader, J.S., Pavlidis, C., Patrinos, G.P., Katsila, T., 2015b. Nutrigenomics: a contro-
Bemben, L.A., Berka, J., Braverman, M.S., Chen, Y.J., Chen, Z., versy. Appl. Transl. Genomics 4, 50e53.
Dewell, S.B., Du, L., Fierro, J.M., Gomes, X.V., Godwin, B.C., Pavlidis, C., Nebel, J.C., Katsila, T., Patrinos, G.P., 2016. Nutrigenomics
He, W., Helgesen, S., Ho, C.H., Irzyk, G.P., Jando, S.C., 2.0: the need for ongoing and independent evaluation and synthesis of
Alenquer, M.L., Jarvie, T.P., Jirage, K.B., Kim, J.B., Knight, J.R., commercial nutrigenomics tests’ scientific knowledge base for
Lanza, J.R., Leamon, J.H., Lefkowitz, S.M., Lei, M., Li, J., responsible innovation. OMICS 20, 65e68.
Lohman, K.L., Lu, H., Makhijani, V.B., McDade, K.E., Petricoin, E.F., Zoon, K.C., Kohn, E.C., Barrett, J.C., Liotta, L.A., 2002.
McKenna, M.P., Myers, E.W., Nickerson, E., Nobile, J.R., Plant, R., Clinical proteomics: translating benchside promise into bedside re-
Puc, B.P., Ronan, M.T., Roth, G.T., Sarkis, G.J., Simons, J.F., ality. Nat. Rev. Drug. Discov. 1, 683e695.
Simpson, J.W., Srinivasan, M., Tartaro, K.R., Tomasz, A., Pirastu, M., Kan, Y.W., Cao, A., Conner, B.J., Teplitz, R.L., Wallace, R.B.,
Vogt, K.A., Volkmer, G.A., Wang, S.H., Wang, Y., Weiner, M.P., 1983. Prenatal diagnosis of beta-thalassemia. Detection of a single
Yu, P., Begley, R.F., Rothberg, J.M., 2005. Genome sequencing in nucleotide mutation in DNA. N. Engl. J. Med. 309, 284e287.
microfabricated high-density picolitre reactors. Nature 437, 376e380. Pisanu, C., Tsermpini, E.E., Mavroidi, E., Katsila, T., Patrinos, G.P.,
Mashal, R.D., Koontz, J., Sklar, J., 1995. Detection of mutations by Squassina, A., 2014. Assessment of the pharmacogenomics educa-
cleavage of DNA heteroduplexes with bacteriophage resolvases. Nat. tional environment in Southeast Europe. Public Health Genomics 17,
Genet. 9, 177e183. 272e279.
Mizzi, C., Mitropoulou, C., Mitropoulos, K., Peters, B., Agarwal, M.R., Potamias, G., Lakiotaki, K., Katsila, T., Lee, M., Topouzis, S.,
van Schaik, R.H., Drmanac, R., Borg, J., Patrinos, G.P., 2014. Cooper, D.N., Patrinos, G.P., 2014. Deciphering next-generation
Personalized pharmacogenomics profiling using whole genome pharmacogenomics: an information technology perspective. Open
sequencing. Pharmacogenomics 15, 1223e1234. Biol. 4, 140071.
Mullis, K.B., Faloona, F.A., 1987. Specific synthesis of DNA in vitro via a Reydon, T.A., Kampourakis, K., Patrinos, G.P., 2012. Genetics, genomics
polymerase-catalyzed chain reaction. Methods Enzymol. 155, 335e350. and society: the responsibilities of scientists for science communica-
Myers, R.M., Larin, Z., Maniatis, T., 1985a. Detection of single base tion and education. Per. Med. 9, 633e643.
substitutions by ribonuclease cleavage at mismatches in RNA: DNA Robinson, W.H., DiGennaro, C., Hueber, W., Haab, B.B., Kamachi, M.,
duplexes. Science 230, 1242e1246. Dean, E.J., Fournel, S., Fong, D., Genovese, M.C., de Vegvar, H.E.,
Myers, R.M., Lumelsky, N., Lerman, L.S., Maniatis, T., 1985b. Detection Skriner, K., Hirschberg, D.L., Morris, R.I., Muller, S., Pruijn, G.J.,
of single base substitutions in total genomic DNA. Nature 1985 (313), van Venrooij, W.J., Smolen, J.S., Brown, P.O., Steinman, L.,
495e498. Utz, P.J., 2002. Autoantigen microarrays for multiplex characteriza-
Myers, R.M., Maniatis, T., Lerman, L.S., 1987. Detection and localization tion of autoantibody responses. Nat. Med. 8, 295e301.
of single base changes by denaturing gradient gel electrophoresis. Roche, M.I., Berg, J.S., 2015. Incidental findings with genomic testing:
Methods Enzymol. 155, 501e527. implications for genetic counseling practice. Curr. Genet. Med. Rep.
Orita, M., Iwahana, H., Kanazawa, H., Hayashi, K., Sekiya, T., 1989. 3 (4), 166e176.
Detection of polymorphisms of human DNA by gel electrophoresis as Ronaghi, M., Uhlen, M., Nyren, P., 1998. A sequencing method based on
single-strand conformation polymorphisms. Proc. Natl. Acad. Sci. real-time pyrophosphate. Science 281, 363e365.
U.S.A. 86, 2766e2770. Saiki, R.K., Bugawan, T.L., Horn, G.T., Mullis, K.B., Erlich, H.A., 1986.
Orkin, S.H., Kazazian Jr., H.H., Antonarakis, S.E., Goff, S.C., Analysis of enzymatically amplified beta-globin and HLA-DQ alpha
Boehm, C.D., Sexton, J.P., Waber, P.G., Giardina, P.J., 1982. Linkage DNA with allele-specific oligonucleotide probes. Nature 324, 163e166.
of beta-thalassaemia mutations and beta-globin gene polymorphisms Saiki, R.K., Gelfand, D.H., Stoffel, S., Scharf, S.J., Higuchi, R.,
with DNA polymorphisms in human betaglobin gene cluster. Nature Horn, G.T., Mullis, K.B., Erlich, H.A., 1988. Primer-directed enzy-
296, 627e631. matic amplification of DNA with a thermostable DNA polymerase.
Orkin, S.H., Markham, A.F., Kazazian Jr., H.H., 1983. Direct detection of Science 239, 487e491.
the common Mediterranean beta-thalassemia gene with synthetic Saiki, R.K., Scharf, S., Faloona, F., Mullis, K.B., Horn, G.T., Erlich, H.A.,
DNA probes. An alternative approach for prenatal diagnosis. J. Clin. Arnheim, N., 1985. Enzymatic amplification of beta-globin genomic
Invest. 71, 775e779. sequences and restriction site analysis for diagnosis of sickle cell
Patrinos, G.P., Baker, D.J., Al-Mulla, F., Vasiliou, V., Cooper, D.N., 2013. anemia. Science 230, 1350e1354.
Genetic tests obtainable through pharmacies: the good, the bad and the Saiki, R.K., Walsh, P.S., Levenson, C.H., Erlich, H.A., 1989. Genetic
ugly. Hum. Genomics 7 (1), 17. analysis of amplified DNA with immobilized sequence-specific
Patrinos, G.P., Giardine, B., Riemer, C., Miller, W., Chui, D.H., oligonucleotide probes. Proc. Natl. Acad. Sci. U.S.A. 86, 6230e6234.
Anagnou, N.P., Wajcman, H., Hardison, R.C., 2004. Improvements in Saleeba, J.A., Ramus, S.J., Cotton, R.G., 1992. Complete mutation
the HbVar database of human hemoglobin variants and thalassemia detection using unlabeled chemical cleavage. Hum. Mutat. 1, 63e69.
mutations for population and sequence variation studies. Nucleic Snyder, S.R., Mitropoulou, C., Patrinos, G.P., Williams, M.S., 2014.
Acids Res. 32, D537eD541. Economic evaluation of pharmacogenomics: a value-based approach
Pauling, L., Itano, H.A., Singer, S.J., Wells, I.C., 1949. Sickle cell anemia, to pragmatic decision making in the face of complexity. Public Health
a molecular disease. Science 110, 543e548. Genomics 17, 256e264.
Molecular Diagnostics: Past, Present, and Future Chapter | 1 11

Squassina, A., Artac, M., Manolopoulos, V.G., Karkabouna, S., Lappa- Johnson, J., Kalush, F., Kline, L., Koduru, S., Love, A., Mann, F.,
Manakou, C., Mitropoulos, K., Manchia, M., del Zompo, M., May, D., McCawley, S., McIntosh, T., McMullen, I., Moy, M.,
Patrinos, G.P., 2010. Translation of genetic knowledge into clinical Moy, L., Murphy, B., Nelson, K., Pfannkoch, C., Pratts, E., Puri, V.,
practice: the expectations and realities of pharmacogenomics and Qureshi, H., Reardon, M., Rodriguez, R., Rogers, Y.H., Romblad, D.,
personalized medicine. Pharmacogenomics 11, 1149e1167. Ruhfel, B., Scott, R., Sitter, C., Smallwood, M., Stewart, E.,
Treisman, R., Orkin, S.H., Maniatis, T., 1983. Specific transcription and Strong, R., Suh, E., Thomas, R., Tint, N.N., Tse, S., Vech, C.,
RNA splicing defects in five cloned beta-thalassaemia genes. Nature Wang, G., Wetter, J., Williams, S., Williams, M., Windsor, S., Winn-
302, 591e596. Deen, E., Wolfe, K., Zaveri, J., Zaveri, K., Abril, J.F., Guigo, R.,
van Ommen, G.J., 2002. The Human genome project and the future of di- Campbell, M.J., Sjolander, K.V., Karlak, B., Kejariwal, A., Mi, H.,
agnostics, treatment and prevention. J. Inherit. Metab. Dis. 25, 183e188. Lazareva, B., Hatton, T., Narechania, A., Diemer, K.,
Venter, J.C., Adams, M.D., Myers, E.W., Li, P.W., Mural, R.J., Muruganujan, A., Guo, N., Sato, S., Bafna, V., Istrail, S., Lippert, R.,
Sutton, G.G., Smith, H.O., Yandell, M., Evans, C.A., Holt, R.A., Schwartz, R., Walenz, B., Yooseph, S., Allen, D., Basu, A.,
Gocayne, J.D., Amanatides, P., Ballew, R.M., Huson, D.H., Baxendale, J., Blick, L., Caminha, M., Carnes-Stine, J., Caulk, P.,
Wortman, J.R., Zhang, Q., Kodira, C.D., Zheng, X.H., Chen, L., Chiang, Y.H., Coyne, M., Dahlke, C., Mays, A., Dombroski, M.,
Skupski, M., Subramanian, G., Thomas, P.D., Zhang, J., Gabor Donnelly, M., Ely, D., Esparham, S., Fosler, C., Gire, H.,
Miklos, G.L., Nelson, C., Broder, S., Clark, A.G., Nadeau, J., Glanowski, S., Glasser, K., Glodek, A., Gorokhov, M., Graham, K.,
McKusick, V.A., Zinder, N., Levine, A.J., Roberts, R.J., Simon, M., Gropman, B., Harris, M., Heil, J., Henderson, S., Hoover, J.,
Slayman, C., Hunkapiller, M., Bolanos, R., Delcher, A., Dew, I., Jennings, D., Jordan, C., Jordan, J., Kasha, J., Kagan, L., Kraft, C.,
Fasulo, D., Flanigan, M., Florea, L., Halpern, A., Hannenhalli, S., Levitsky, A., Lewis, M., Liu, X., Lopez, J., Ma, D., Majoros, W.,
Kravitz, S., Levy, S., Mobarry, C., Reinert, K., Remington, K., Abu- McDaniel, J., Murphy, S., Newman, M., Nguyen, T., Nguyen, N.,
Threideh, J., Beasley, E., Biddick, K., Bonazzi, V., Brandon, R., Nodell, M., Pan, S., Peck, J., Peterson, M., Rowe, W., Sanders, R.,
Cargill, M., Chandramouliswaran, I., Charlab, R., Chaturvedi, K., Scott, J., Simpson, M., Smith, T., Sprague, A., Stockwell, T.,
Deng, Z., Di Francesco, V., Dunn, P., Eilbeck, K., Evangelista, C., Turner, R., Venter, E., Wang, M., Wen, M., Wu, D., Wu, M., Xia, A.,
Gabrielian, A.E., Gan, W., Ge, W., Gong, F., Gu, Z., Guan, P., Zandieh, A., Zhu, X., 2001. The sequence of the human genome.
Heiman, T.J., Higgins, M.E., Ji, R.R., Ke, Z., Ketchum, K.A., Lai, Z., Science 291, 1304e1351.
Lei, Y., Li, Z., Li, J., Liang, Y., Lin, X., Lu, F., Merkulov, G.V., Wheeler, D.A., Srinivasan, M., Egholm, M., Shen, Y., Chen, L.,
Milshina, N., Moore, H.M., Naik, A.K., Narayan, V.A., Neelam, B., McGuire, A., He, W., Chen, Y.J., Makhijani, V., Roth, G.T.,
Nusskern, D., Rusch, D.B., Salzberg, S., Shao, W., Shue, B., Sun, J., Gomes, X., Tartaro, K., Niazi, F., Turcotte, C.L., Irzyk, G.P.,
Wang, Z., Wang, A., Wang, X., Wang, J., Wei, M., Wides, R., Lupski, J.R., Chinault, C., Song, X.Z., Liu, Y., Yuan, Y., Nazareth, L.,
Xiao, C., Yan, C., Yao, A., Ye, J., Zhan, M., Zhang, W., Zhang, H., Qin, X., Muzny, D.M., Margulies, M., Weinstock, G.M., Gibbs, R.A.,
Zhao, Q., Zheng, L., Zhong, F., Zhong, W., Zhu, S., Zhao, S., Rothberg, J.M., 2008. The complete genome of an individual by
Gilbert, D., Baumhueter, S., Spier, G., Carter, C., Cravchik, A., massively parallel DNA sequencing. Nature 452, 872e876.
Woodage, T., Ali, F., An, H., Awe, A., Baldwin, D., Baden, H., Woo, S.L., Lidsky, A.S., Guttler, F., Chandra, T., Robson, K.J., 1983.
Barnstead, M., Barrow, I., Beeson, K., Busam, D., Carver, A., Cloned human phenylalanine hydroxylase gene allows prenatal
Center, A., Cheng, M.L., Curry, L., Danaher, S., Davenport, L., diagnosis and carrier detection of classical phenylketonuria. Nature
Desilets, R., Dietz, S., Dodson, K., Doup, L., Ferriera, S., Garg, N., 306, 151e155.
Gluecksmann, A., Hart, B., Haynes, J., Haynes, C., Heiner, C., Xiao, W., Oefner, P.J., 2001. Denaturing high-performance liquid chro-
Hladun, S., Hostin, D., Houck, J., Howland, T., Ibegwam, C., matography: a review. Hum. Mutat. 17, 439e474.

Vous aimerez peut-être aussi