You are on page 1of 11

Basic Science / THE INTEGRATION OF MOLECULAR DIAGNOSTICS WITH THERAPEUTICS

The Integration of Molecular Diagnostics


With Therapeutics
Implications for Drug Development and Pathology Practice
Jeffrey S. Ross, MD,1,2 and Geoffrey S. Ginsburg, MD, PhD2

Key Words: Personalized medicine; Molecular diagnostics; Pharmacogenomics; Pharmacogenetics; Toxicogenomics; Cancer predisposition;
Productivity

DOI: 10.1092/VMLL66Y5KHQ35KUE

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


Abstract Molecular pathology is in a state of rapid evolution
It is widely anticipated that during the next 5 years featuring continuous technology developments and new clin-
the molecular diagnostic industry will continue to grow ical opportunities for drug selection, predicting efficacy and
at double-digit pace to meet increasing demand for toxicity, and monitoring disease outcome.1-3 The approvals of
personalized medicine. A wide variety of drugs in late trastuzumab (Herceptin, Genentech, South San Francisco,
preclinical and early clinical development are being CA) for the treatment of HER-2/neu overexpressing breast
targeted to disease-specific gene and protein defects that cancer and imatinib (Gleevec, Novartis Pharmaceuticals, East
will require coapproval of diagnostic and therapeutic Hanover, NJ) for the treatment of chronic myelogenous
products by regulatory agencies. An increasingly leukemia featuring a bcr/abl translocation and gastroin-
educated public will demand more information about testinal stromal tumors with selective c-kit oncogene–acti-
their predisposition for serious diseases and how these vating mutations have brought to the diagnostic laboratory an
potential illnesses can be detected in an early stage expanding role for the testing of patients to determine their
when they can be arrested or cured with new therapies eligibility to receive these new therapies.4,5 The introduction
custom-designed for their individual clinical status. To of targeted therapeutics into clinical practice has created
respond to this demand, major pharmaceutical major opportunities for further development of the molecular
companies will partner with diagnostics companies or pathology. As shown in ❚Table 1❚, the molecular diagnostics
develop their own in-house capabilities that will permit industry is a market greater than $3 billion, which currently is
efficient production of more effective and less toxic growing at approximately 25% per year. The worldwide
integrated personalized medicine drug and test products. pharmaceuticals annual market is estimated to exceed $1.1
For clinical laboratories and pathologists, this trillion by 2010. It is widely held that, during the next 5 to 10
integration of diagnostics and therapeutics represents a years, the clinical application of molecular pathology and
major new opportunity to emerge as leaders of the new molecular diagnostics will further revolutionize the drug
medicine, guiding the selection, dosage, route of discovery and development process; customize the selection,
administration, and multidrug combinations and dosing, and route of administration of existing and new thera-
producing increased efficacy and reduced toxicity of peutic agents; and truly personalize medical care.6,7
pharmaceutical products. Many so-called blockbuster drugs show only limited
efficacy in as many as 70% of treated patients8 ❚Table 2❚. The
current roster of phenotypically derived drugs that often treat
only the symptoms of diseases but do not cure them is no
longer acceptable to the public. Moreover, adverse drug reac-
tions caused by the failure to predict toxicity in individuals
and toxic drug-drug interactions now account for 100,000

26 Am J Clin Pathol 2003;119:26-36 © American Society for Clinical Pathology


26 DOI: 10.1092/VMLL66Y5KHQ35KUE
Basic Science / REVIEW ARTICLE

❚Table 1❚
Estimated Worldwide Molecular Diagnostics Markets*

2000 2005 2010

By diagnostic technique
DNA probes 240 400 850
Polymerase chain reaction–based 1,450 2,200 4,350
Fluorescence in situ hybridization 600 850 2,350
Arrays 400 1,300 3,300
Others 450 750 1,150
Total 3,140 5,500 12,000
By clinical setting
Infections 2,080 2,920 5,100
Cancer 337 900 1,900
Genetic tests 320 792 2,007
Food supply testing 110 198 400
Others 293 690 1,593
Total 3,140 5,500 11,000
* Modified from Jain KK. Molecular Diagnostics: Technologies, Markets, Companies. Basel, Switzerland: Jain PharmaBiotech Publications; 2002. Data are given in millions of
US dollars.

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


❚Table 2❚ measurements of drug efficacy and risk of toxic effects, and
Examples of the Variable Efficacy of Phenotypically the monitoring of the illness until the final disease outcome is
Prescribed Drugs
known ❚Figure 1❚. Emerging genomic and proteomic tech-
Frequency of Absent or nologies and information are resulting in the molecular
Drug Class Incomplete Efficacy (%) subclassification of disease as the basis for diagnosis, deter-
Angiotensin-converting enzyme inhibitors 10-30 mining the prognosis, and therapeutic selection. In the past,
Beta blockers 15-25 medicine relied on nonspecific clinical signs for diagnosis
Antidepressants 20-50
Statins 30-70 and empiric treatment strategies. For example, expanded
Beta2-agonists 40-70 knowledge of the molecular basis of cancer has shown that
significant differences in gene expression patterns can guide
therapy for a variety of solid tumors and hematologic malig-
patient deaths, 2 million hospitalizations, and $100 billion in nant neoplasms.13,14 In cardiovascular disease, genetic hetero-
health care costs in the United States every year.9-12 As many geneity has been identified in the long QT syndrome, and
as 20% to 40% of people receiving pharmaceutical agents choice of therapy (sodium channel blocker vs K+ channel
may be receiving the wrong drug.9-11 The discovery of the blocker vs beta blocker) now is determined by the genetic
human genome and the subsequent expansion of proteomics etiology of the syndrome.15-17
research combined with emerging technologies such as func-
tional imaging, biosensors, and sophisticated computational
biology are producing unprecedented changes in today’s
Goals of Personalized Medicine
health care. A variety of new approaches to drug use have
introduced genetically prescribed medications ❚Table 3❚. The ultimate goals of personalized medicine are to
Moreover, modern medicine now is driven by rapid commu- take advantage of a molecular understanding of disease to
nication and electronic information sharing, which has optimize drug development and to direct preventive
created more informed and demanding consumers. resources and therapeutic agents at the right population of
people while they are still well. The goals of personalized
medicine in drug development include the following: (1)
the selection of optimal drug targets; (2) the selection of
Personalized Medicine
optimal drug dosage; (3) the selection and monitoring of
Personalized medicine includes the concepts that for a patients for shorter, less expensive, advanced clinical
given disease, the rate of progression of the disease for each trials; (4) the ability to predict which individuals will
person is unique and each person responds in a unique way to respond to drugs at high rates and which individuals will
drugs.12 In its broadest sense, personalized medicine includes be less likely to have toxic effects; (5) the reduction of the
the detection of disease predisposition, screening and early overall cost of drug development and an increase in drug
disease diagnosis, assessment of prognosis, pharmacogenomic value; and (6) ultimate improvement and provision of

© American Society for Clinical Pathology Am J Clin Pathol 2003;119:26-36 27


2 DOI: 10.1092/VMLL66Y5KHQ35KUE 27
Ross and Ginsburg / THE INTEGRATION OF MOLECULAR DIAGNOSTICS WITH THERAPEUTICS

❚Table 3❚
Genetically Prescribed Medications

Target Drug Disease Action

Cytochrome P-450s Warfarin Coagulation disorders Toxicity


TPMT Mercaptopurine Cancer Toxicity
CETP HMG-CoA reductase inhibitors Atherosclerosis Efficacy
APOE-4 Tacrine Alzheimer Efficacy
HER-2 receptor Trastuzumab Cancer Efficacy
bcr-abl Imatinib Cancer Efficacy
Estrogen receptor Hormone replacements Osteoporosis Efficacy
BRCA2 Tamoxifen Cancer Efficacy
Transcriptional profiles Taxanes, anthracyclines, platinins Ovarian cancer, leukemia, breast cancer Efficacy

APOE-4, apolipoprotein E, epsilon 4; BRCA, breast cancer; CETP, cholesteryl ester transfer protein; HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A; TPMT, thiopurine S-
methyltransferase.

more effective health care for all individuals whether well Disease outcome

Disease Management

y
or in the early or late stages of illness.

og

Symptom Severity
ol
Genetic variants can be used to predict the predisposi- Monitoring

th

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


pa
tion of a person for future disease development such as

ar
Pharmacogenomic

ul
cancer ❚Table 4❚. Preventive therapies currently in use for a

ec
ol
Clinical

M
variety of diseases include selective estrogen receptor modu- Prognostic
symptoms
lators for patients at risk for breast cancer 18 and
Normal Screening Subclinical
osteoporosis19 and statins for people at risk for developing
health disease process
coronary artery disease.20 Predisposition

Time

Productivity in the Pharmaceutical


❚Figure 1❚ Personalized medicine. Personalized medicine
Industry and Personalized Medicine
includes the detection of disease predisposition, screening
By applying the principles of personalized medicine, it and early disease diagnosis, prognosis assessment,
is possible to enhance substantially the productivity of drug pharmacogenomic measurements of drug efficacy and risk of
discovery and development. Through the identification of the toxic effects, and the monitoring of the illness until the final
right gene, the right pathway, and the right target in the disease outcome is known.
pathway, the right drug can be developed to treat the right
patient. Estimated potential reductions by 20% in the
number of new compounds tested in phase 2 and 3 human known as single nucleotide polymorphisms (SNPs) have
clinical trials, by 50% in the number of patients in phase 2 become available for genotyping and phenotyping stud-
trials, by 10% in the number of patients in phase 3 trials, and ies.21-24 The use of a variety of techniques for cloning and
by 20% in the length of phase 3 trials are well within reach.8 sequencing and an evolving number of informatics
The net result of biomarker-assisted drug development is an approaches leading to the successful mining of high-quality
overall reduction of compound failures in clinical trials and sequence variations have revealed numerous loci that seem
on the market and a cost savings of perhaps as much as $500 to have substantial potential to generate clinically useful data
million for each drug launched. for patient management. On a regular basis, novel geno-
typing strategies are emerging that use a variety of tech-
niques, including oligonucleotide genomic arrays,21,22 gel
and flow cytometry, classic gene sequencing, mass spec-
Pharmacogenetics and Single
troscopy,23 and microarray or gene chips,24 all designed to
Nucleotide Polymorphisms
increase the rate of data generation and analysis. In 1999, 10
An individual’s response to a drug is the complex inter- companies and the Wellcome Trust formed a consortium to
action of both genetic and nongenetic factors. Genetic vari- discover clinically relevant SNPs. To date, more than 2.1
ants in the drug target itself, disease pathway genes, and drug million SNPs have been deposited into the SNP Consor-
metabolizing enzymes all may be used as predictors of drug tium’s public database (http://www.snp.cshl.org). A high-
efficacy or toxicity. More than 1 million genetic markers resolution SNP map will expedite the identification of genes

28 Am J Clin Pathol 2003;119:26-36 © American Society for Clinical Pathology


28 DOI: 10.1092/VMLL66Y5KHQ35KUE
Basic Science / REVIEW ARTICLE

❚Table 4❚
Cancer Predisposition and Tumor Suppressor Genes

Gene Name Designation Location Disease Association

Adenomatous polyposis coli APC 5q21 Familial polyposis coli


Breast cancer 1 BRCA1 17q21 Familial breast cancer
Breast cancer 2 BRCA2 13q12 Familial breast cancer
E-cadherin CDH1 16q22 Familial stomach cancer; lobular breast cancer (also
determining prognosis in epithelial cancers)
Cyclin-dependent kinase inhibitor 1C CDKN1C 16q22 Beckwith-Wiedemann syndrome; Wilms tumor;
rhabdomyosarcoma
p16 Cyclin-dependent kinase inhibitor 2A CDKN2A 11p15 Familial melanoma; bladder cancer; other epithelial tumors
(MTS 1; INK4A)
Mitogen-activated protein kinase 4 MAP2K4 17p11 Pancreatic, breast, colorectal cancer
Multiple endocrine neoplasia type 1 (type 2 MEN1 11q13 Pituitary adenoma; parathyroid adenoma; islet cell
associated with the ret oncogene) carcinoma; carcinoids
Hereditary nonpolyposis colorectal cancer type 1 hMSH2 2p22 Colorectal cancer; ovarian cancer; endometrial cancer
Hereditary nonpolyposis colorectal cancer type 1 hMLH1 3p21 Colorectal cancer; ovarian cancer; endometrial cancer
Neurofibromatosis type 1 NF1 17q11 Neurofibroma; glioma; pheochromocytoma; acute
myelogenous leukemia
Neurofibromatosis type 2 NF2 22q12 Schwannoma; meningioma; ependymoma
Phosphatase and tensin homologue PTEN 10q23 Cowden syndrome; glioma; prostate cancer; endometrial cancer
Retinoblastoma RB1 13q14 Retinoblastoma; osteosarcoma; small cell lung cancer;

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


breast cancer
Serine/threonine kinase 11 STK11 19p13 Peutz-Jeghers syndrome
TP53 p53 17p13 Li-Fraumeni syndrome; inactive in >50% of human malignant
neoplasms
Tuberous sclerosis 1 TSC1 (KIAA023) 9q34 Hamartoma; renal cell carcinoma; angiomyolipoma
Tuberous sclerosis 2 TSC2 16p13 Hamartoma; renal cell carcinoma; angiomyolipoma
von Hippel-Lindau VHL 3p26 Hemangioma; renal cell carcinoma; pheochromocytoma
Wilms tumor 1 WT1 11p13 Genitourinary dysplasia; familial Wilms tumor

for complex diseases such as asthma, diabetes mellitus, Prediction of Drug Efficacy
atherosclerosis, and psychiatric disorders. In oncology, SNP The application of genotyping strategies to predict anti-
technology has focused on detecting the predisposition for cancer drug efficacy has emerged in a variety of clinical
cancer, predicting toxic responses to drugs, and selecting the settings.35-39 Genotype resistance testing of HIV isolates has
best individual and combination anticancer drugs. demonstrable clinical usefulness and provides a way to assist
therapeutic decision making for patients whose HIV RNA
Cancer Predisposition Testing levels are rising.40 In colorectal cancer, pretreatment geno-
SNP genotyping and gene sequencing (see “Predisposi- typing on peripheral blood samples is being used to select
tion, Screening, and Early Diagnosis of Disease”) have therapy based on the prediction of resistance associated with
revealed a variety of familiar cancer predisposition certain genetic polymorphisms.39
syndromes (Table 4) based on single and multiple gene vari-
ants.24-27 Genotyping has been introduced widely for the
detection of familial cancers of the breast, ovary, and colon;
Pharmacogenomics
melanoma; and multiple endocrine neoplasia.26,28-30
Pharmacogenomics can best be defined as the applica-
Prediction of Drug Toxicity tion of whole genome technologies (eg, gene and protein
One of the earliest applications of SNP genotyping in expression data) for the prediction of the sensitivity or
cancer management was the discovery of variations in drug resistance of an individual’s disease to a single drug or
metabolism associated with genomic variations in drug- group of drugs.41,42
metabolizing enzymes such as the cytochrome system.31-33
Other detoxification pathways associated with SNP-based Transcriptional Profiling and Genomic Microarrays
variations in drug metabolism have been described,32-34 but, The development of printed and spotted genomic
in comparison with the cytochrome gene system, these microarrays has permitted the rapid accumulation of new
markers have not achieved substantial clinical usefulness. information about gene mutation and expression in human
The potential clinical value of the pharmacogenetics malignant neoplasms.43-50 Microarrays can be used to deter-
approach for predicting drug toxicity will be revealed as mine gene mutations and SNPs and to provide rapid
more candidate polymorphisms are discovered. screening information about messenger RNA expression. In

© American Society for Clinical Pathology Am J Clin Pathol 2003;119:26-36 29


2 DOI: 10.1092/VMLL66Y5KHQ35KUE 29
Ross and Ginsburg / THE INTEGRATION OF MOLECULAR DIAGNOSTICS WITH THERAPEUTICS

transcriptional profiling, the relative expression levels of of anticancer drugs has been used as a guide to anticancer
messenger RNA of both characterized and novel genes drug therapy. 50-56 By using cDNA microarrays, several
from a tumor can be compared with the gene expression groups have reported their success at discovering gene
status of a reference sample. Reference samples can expression that can be linked to resistance and responsive-
include cell lines, normal tissues, and cancer precursor ness to standard-of-care chemotherapy.57,58 During the next
lesions. 43-50 Transcriptional profiling has the ability to several years, the ability of this approach to personalize the
generate hundreds of thousands of data points, and it treatment of newly diagnosed cancer patients with individu-
requires sophisticated and complex information systems for alized selection and dosage of chemotherapeutic agents will
accurate and useful data analysis. Complementary DNA be tested on a large scale.
(cDNA) microarrays were introduced during the mid-
1990s51 and have achieved widespread use for the expres-
sion profiling of human clinical samples and in drug and
Pharmacodynamics
biomarker target discovery. The microarray technique has
generated a wealth of new information in the fields of In addition to the pharmacogenetic and pharmacoge-
leukemia and lymphoma and solid tumor classification, nomic tests, a series of bioassays, gene expression profiles,
drug and biomarker target discovery, and pharmacoge- and tissue-based biomarkers have emerged to guide the
nomic drug efficacy testing43-50 ❚Image 1❚. Oligonucleotide dosage, timing, and route of administration for novel thera-

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


microarrays have been used mostly to detect SNPs and peutics. These tests are used to speed the preclinical and early
individual gene point mutations and for profiling differ- clinical development of drugs by enhancing the achievement
ences in gene expression between samples from human of the ideal therapeutic dose while avoiding dose-related toxic
tumors and reference test tissues. The oligonucleotide effects. Recent examples of these novel assays include the
microarray technique is, however, well-regarded as a blood-based bioassay of the proteasome designed to guide the
method of automated and robotic sampling of the genome use of the proteasome inhibitor MLN-341 in multiple
in search of SNPs and other clinically relevant polymorphisms.52 myeloma and solid tumors59-61 and the immunohistochemical
determination of the epidermal growth factor receptor
Transcriptional Profiling and Pharmacogenomics (EGFR) target and cell proliferation markers in skin biopsy
The hierarchical clustering technique of data analysis from specimens of patients receiving the anti-EGFR small mole-
transcriptional profiling of clinical samples known to have cule tyrosine kinase inhibitor geftinib (Iressa, AstraZeneca
responded or been resistant to a single agent or combination Pharmaceuticals, Manchester, England).62

❚Image 1❚ Breast cancer complementary DNA (cDNA) microarray results evaluated by hierarchical gene cluster analysis for defining
specific gene expression signatures. Hierarchical clustering algorithm permits the clustering of individual tumor profiles on the
basis of their similarities to their coexpression with the estrogen receptor alpha gene. Each row represents a tumor and each
column a single gene. Red indicates up-regulation; green, down-regulation; and black, no change in relative gene expression.

30 Am J Clin Pathol 2003;119:26-36 © American Society for Clinical Pathology


30 DOI: 10.1092/VMLL66Y5KHQ35KUE
Basic Science / REVIEW ARTICLE

Toxicogenomics patients with chronic myelogenous leukemia or activating


mutations in the c-kit tyrosine kinase receptor in patients
Toxicogenomics is the study of gene expression with gastrointestinal stromal tumors for the selection of the
patterns using high-throughput microarrays, automated small molecule drug, imatinib.5,70,71 A substantial number
reverse transcriptase–polymerase chain reaction, nuclear of small molecule drugs in current clinical trials are targeted
magnetic resonance, and proteomic strategies designed to to a variety of growth factor receptors, angiogenesis
detect up-regulation and down-regulation of genes associ- promoters, cell cycle regulators, and invasion or metastasis
ated with the risk of drug toxic effects.63-66 Toxicogenomic biomarkers.72-74 Similarly, a variety of antitumor antibodies
markers for adverse effects may influence selection and have been targeted specifically to cell surface antigens and
optimization of lead compounds before human studies. For designed to deliver cytotoxic radioisotopes75 and cellular
example, inherited forms of the long QT syndrome can be toxins.76 During the next 5 years, the impact of the targeted
caused by high-affinity drug blockade associated with approach in which diagnostic tests guide the clinical selec-
mutations in the HERG potassium channel regulatory tion of anticancer drugs will become known.
gene.17,67 The limitations and uncertainties of gene expres-
sion profiling associated with data mining constraints on
mechanistic and predictive toxicology have been empha-
Integrated Diagnostics and Therapeutics
sized, and the clinical value of toxicogenomics is mostly
in Nonmalignant Diseases

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


unrealized.66
A wide variety of existing and novel diagnostic tests
have been integrated into the management strategy of many
major nonmalignant diseases. A number of groups have
Targeted Therapies
studied patients with rheumatoid arthritis to learn whether
The integration of diagnostics and therapeutics is real- germline-based SNP detection or biomarker assays
ized in the discovery and development of targeted therapies. performed on serum or synovial fluid could predict the
Targeted therapy has emerged in new cancer treatment development of erosive disease and guide the selection of
strategies as evidenced in the approvals of trastuzumab and therapy.77,78 Patients with inflammatory bowel disease have
imatinib. 4,5 The features of the ideal anticancer target been tested for biomarkers of disease severity as a method
designed to maximize efficacy and minimize toxic effects of selecting local vs systemic drug therapy.79,80 In addition to
include the following: (1) The target is crucial for the exis- classic measurements of known risk factors, new strategies
tence of the malignant phenotype, not significantly for predicting the risk of early-onset coronary atheroscle-
expressed in vital organs and tissues, a biologically relevant rosis have emerged that use well-established traditional
molecular feature, measurable in readily obtained clinical biomarkers such as serum C-reactive protein levels in new
samples in a reproducible manner, and clearly correlated ways81 and novel biomedical imaging and molecular diag-
with clinical outcome. (2) Interruption of, interference with, nostic tests.82,83 Similarly, new integrations of genotyping
or inhibition of the target yields a clinical response in a and molecular diagnostics are widening the knowledge base
substantial proportion of patients whose tumors express the and influencing the management of people with diabetes
target. (3) Responses in patients whose tumors do not mellitus.84 Finally, the widest integration of molecular diag-
express the target are minimal. nostics in current clinical practice is in the selection and
In 1999, the US Food and Drug Administration monitoring of therapy for infectious diseases. Viral load
approvals of trastuzumab and HercepTest (DAKO, Carpin- testing for people infected with HIV or the hepatitis C virus
teria, CA) for patients with HER-2/neu overexpressing is the most frequently performed nucleic acid–based test in
breast cancers marked the first simultaneous regulatory molecular diagnostics laboratories.85 In addition, the use of
review of a targeted therapeutic with its companion diag- antiretroviral drug resistance testing has become the stan-
nostic test.4,13,68,69 A variety of modifications of the HER- dard of care for managing patients with AIDS.86,87
2/neu testing platform have been introduced and are
designed to achieve the best prediction of trastuzumab
response ❚Image 2❚. This integration of drug selection based
Predisposition, Screening, and Early
on a diagnostic test result remains among the foremost of
Diagnosis of Disease
targeted therapy strategies currently available for cancer
patients. Another recent example of targeted therapy in In addition to their direct roles in drug development
oncology in which a diagnostic test specifically directs and clinical use, diagnostic tests are integrated with thera-
therapy is the detection of either the bcr/abl translocation in peutics in a variety of additional applications. Molecular

© American Society for Clinical Pathology Am J Clin Pathol 2003;119:26-36 31


3 DOI: 10.1092/VMLL66Y5KHQ35KUE 31
Ross and Ginsburg / THE INTEGRATION OF MOLECULAR DIAGNOSTICS WITH THERAPEUTICS

A B

❚Image 2❚ HER-2/neu testing in breast cancer. A, HER-2/neu

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


C
gene amplification detected by fluorescence in situ hybridization
(Ventana Inform System, Ventana, Tucson, AZ). B, Immuno-
histochemical analysis using the HercepTest system (DAKO,
Carpinteria, CA) with continuous membranous 3+ positive
immunostaining for HER-2/neu protein (×400). C, HER-2/neu
gene amplification detected by chromogenic in situ
hybridization (Zymed System, Zymed, South San Francisco,
CA) (×400).

diagnostic tests are performed widely to detect cancer 150


Relative Fluorescence

130
predisposition,28-30,88 to screen for malignant and nonmalig- 110
nant diseases,89-92 and to facilitate diagnosis of early and 90
70
recurrent disease ❚Figure 2❚ and ❚Image 3❚ when therapy can 50
be most effective.93,94 High-throughput proteomic profiling 30
10
has emerged as a potential breakthrough in early disease –10
–30
detection and therapy monitoring.95-98 A recent serum-based – 50
1 5 10 15 20 25 30 35 40
study using the surface-enhanced laser desorption ionization
CT Number of PCR Cycles
technique combined with self-learning, computer pattern
recognition software successfully detected early stage
ovarian cancer with high sensitivity and specificity. 99 ❚Figure 2❚ Micrometastasis detection of breast cancer in a
Biomedical and function imaging including computed sentinel axillary lymph node biopsy specimen. Real-time
tomography, magnetic resonance imaging, and positron polymerase chain reaction (PCR; TaqMan, Applied
emission tomography scanning also have contributed major Biosystems, Foster City, CA) detection of cytokeratin 19
advances in the early detection of disease ❚Image 4❚ and in messenger RNA after 25 cycles of amplification (red)
the monitoring of therapeutic response.100 normalized to internal control dye (blue).

32 Am J Clin Pathol 2003;119:26-36 © American Society for Clinical Pathology


32 DOI: 10.1092/VMLL66Y5KHQ35KUE
Basic Science / REVIEW ARTICLE

A B

❚Image 3❚ Micrometastasis detection of breast cancer in a sentinel axillary lymph node biopsy specimen. A, Section of half of a

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


lymph node showing no evidence of tumor cells (H&E, ×400). B, In the same lymph node, a single aggregate of 2 malignant
cells beneath the surface in the subcapsular sinusoid is shown (immunohistochemical stain for cytokeratin 19, ×200).

Day 0 Day 2 Day 4

❚Image 4❚ Metastatic prostate cancer in cervical lymph nodes. Radiolabeled indium scan using the J591 monoclonal antibody,
which detects the external domain of prostate-specific membrane antigen. The scan confirms that the initial clinical finding of a
neck mass was a cervical metastasis from prostate cancer. This patient subsequently had a significant (>50%) reduction in
serum prostate-specific antigen level in response to a single dose of yttrium 90 cytotoxic radio-conjugate of J591.

Conclusion of these challenges are being addressed. As patients continue


to take on more and more of the burden of their own health
The traditional trial-and-error practice of medicine is and well-being, educational forums must be developed for
eroding progressively in favor of more precise marker-assisted patients and providers alike to understand the complex
diagnosis and safer and more effective molecularly guided nature of the genomic and proteomic information that is
treatment of disease. For the pharmaceutical industry, there driving biomarker-based drug development and the future
seems to be an equally desirable outcome of this approach: introduction of integrated diagnostics and therapeutics.
increased efficiency, productivity, and product lines. The
diagnostics industry has an unprecedented opportunity for From the 1Department of Pathology and Laboratory Medicine,
integration, increased value, and commercial opportunities Albany Medical College, Albany, NY; and 2Division of Molecular
for molecularly derived tests. The realization of the integra- Medicine, Millennium Pharmaceuticals, Cambridge, MA.
tion of diagnostics with therapeutics and the transition to Address reprint requests to Dr Ross: Dept of Pathology and
personalized medicine are not without challenges, yet many Laboratory Medicine, Albany Medical College, Albany, NY 12208.

© American Society for Clinical Pathology Am J Clin Pathol 2003;119:26-36 33


3 DOI: 10.1092/VMLL66Y5KHQ35KUE 33
Ross and Ginsburg / THE INTEGRATION OF MOLECULAR DIAGNOSTICS WITH THERAPEUTICS

Acknowledgments: We are indebted to F. Symmans, MD, and 18. Jordan VC. Progress in the prevention of breast cancer:
L. Pustzai, MD, M.D. Anderson Cancer Center, Houston, TX; and concept to reality. J Steroid Biochem Mol Biol. 2000;74:269-277.
N. Bander, MD, New York Presbyterian Hospital, New York, NY, 19. Doren M, Samsioe G. Prevention of postmenopausal
for their collaborative efforts. osteoporosis with oestrogen replacement therapy and
associated compounds: update on clinical trials since 1995.
Hum Reprod Update. 2000;6:419-426.
20. Crouch MA. Effective use of statins to prevent coronary heart
References disease. Am Fam Physician. 2001;63:309-320.
1. Keesee SK. Molecular diagnostics: impact upon cancer 21. Taylor JG, Choi EH, Foster CB, et al. Using genetic variation
detection. Expert Rev Mol Diagn. 2002;2:91-92. to study human disease. Trends Mol Med. 2001;7:507-512.
2. Poste G. Molecular diagnostics: a powerful new component of 22. Carlson CS, Newman TL, Nickerson DA. SNPing in the
the healthcare value chain. Expert Rev Mol Diagn. 2001;1:1-5. human genome. Curr Opin Chem Biol. 2001;5:78-85.
3. Leonard DG. The present and future of molecular diagnostics. 23. Griffin TJ, Smith LM. Single-nucleotide polymorphism
Mol Diagn. 2001;6:71-72. analysis by MALDI TOF mass spectrometry. Trends Biotechnol.
4. Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety 2000;18:77-84.
of trastuzumab as a single agent in first-line treatment of 24. Kallioniemi OP. Biochip technologies in cancer research. Ann
HER2-overexpressing metastatic breast cancer. J Clin Oncol. Med. 2001;33:142-147.
2002;20:719-726. 25. Relling MV, Dervieux T. Pharmacogenetics and cancer
5. O’Dwyer ME, Druker BJ. STI571: an inhibitor of the BCR- therapy. Nat Rev Cancer. 2001;1:99-108.
ABL tyrosine kinase for the treatment of chronic 26. Borg A. Molecular and pathological characterization of

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


myelogenous leukaemia. Lancet Oncol. 2000;1:207-211. inherited breast cancer. Semin Cancer Biol. 2001;11:375-385.
6. Amos J, Patnaik M. Commercial molecular diagnostics in the 27. Renegar G, Rieser P, Manasco P. Pharmacogenetics: the Rx
US: the Human Genome Project to the clinical laboratory. perspective. Expert Rev Mol Diagn. 2001;1:255-263.
Hum Mutat. 2002;19:324-333.
28. Jass JR. Familial colorectal cancer: pathology and molecular
7. Bottles K. A revolution in genetics: changing medicine, characteristics. Lancet Oncol. 2000;1:220-226.
changing lives. Physician Exec. 2001;27:58-63.
29. Weber W, Estoppey J, Stoll H. Familial cancer diagnosis.
8. Halapi E, Hakonarson H. Advances in the development of Anticancer Res. 2001;21:3631-3635.
genetic markers for the diagnosis of disease and drug response.
Expert Rev Mol Diagn. 2002;2:411-421. 30. Calzone KA, Biesecker BB. Genetic testing for cancer
predisposition. Cancer Nurs. 2002;25:15-25.
9. Bordet R, Gautier S, Le Louet H, et al. Analysis of the direct
cost of adverse drug reactions in hospitalised patients. Eur J 31. Ingelman-Sundberg M. Genetic susceptibility to adverse
Clin Pharmacol. 2001;56:935-941. effects of drugs and environmental toxicants: the role of the
CYP family of enzymes. Mutat Res. 2001;482:11-19.
10. Suh DC, Woodall BS, Shin SK, et al. Clinical and economic
impact of adverse drug reactions in hospitalized patients. Ann 32. Ingelman-Sundberg M. Pharmacogenetics: an opportunity for
Pharmacother. 2000;34:1373-1379. a safer and more efficient pharmacotherapy. J Intern Med.
2001;250:186-200.
11. du Souich P. In human therapy, is the drug-drug interaction or
the adverse drug reaction the issue? Can J Clin Pharmacol. 33. Steimer W, Potter JM. Pharmacogenetic screening and
2001;8:153-161. therapeutic drugs. Clin Chim Acta. 2002;315:137-155.
12. Ginsburg GS, McCarthy JJ. Personalized medicine: 34. Roses AD. Pharmacogenetics. Hum Mol Genet. 2001;10:2261-
revolutionizing drug discovery and patient care. Trends 2267.
Biotechnol. 2001;19:491-496. 35. Diasio RB, Johnson MR. The role of pharmacogenetics and
13. Ross JS, Fletcher JA. The HER-2/neu oncogene in breast pharmacogenomics in cancer chemotherapy with 5-
fluorouracil. Pharmacology. 2000;61:199-203.
cancer: prognostic factor, predictive factor, and target for
therapy. Oncologist. 1998;3:237-252. 36. Gold B. DNA genotyping. Adv Clin Chem. 2001;36:171-234.
14. Morgan GJ, Pratt G. Modern molecular diagnostics and the 37. Linder MW, Valdes R Jr. Genetic mechanisms for variability
management of haematological malignancies. Clin Lab in drug response and toxicity. J Anal Toxicol. 2001;25:405-413.
Haematol. 1998;20:135-141. 38. Danesi R, De Braud F, Fogli S, et al. Pharmacogenetic
15. Zareba W, Moss AJ, Schwarts PJ, et al, for the International determinants of anti-cancer drug activity and toxicity. Trends
Long Q-T Syndrome Registry Research Group. Influence of Pharmacol Sci. 2001;22:420-426.
genotype on clinical course of the long-QT syndrome. N Engl 39. Pullarkat ST, Stoehlmacher J, Ghaderi V, et al. Thymidylate
J Med. 1998;339:960-965. synthase gene polymorphism determines response and toxicity
16. Shimizu W, Antzelevitch C. Sodium channel block with of 5-FU chemotherapy. Pharmacogenomics J. 2001;1:65-70.
mexiletine is effective in reducing dispersion of repolarization 40. Durant J, Clevenbergh P, Halfon P, et al. Drug-resistance
and preventing torsade de pointes in LQT2 as well as LQT3 genotyping in HIV-1 therapy: the VIRADAPT randomised
models of the long-QT syndrome. Circulation. 1997;96:2038- controlled trial. Lancet. 1999;353:2195-2199.
2047. 41. Rusnak JM, Kisabeth RM, Herbert DP, et al.
17. Schwartz PJ, Priori SG, Locati EH, et al. Long QT syndrome Pharmacogenomics: a clinician’s primer on emerging
patients with mutations of the SCN5A and HERG genes technologies for improved patient care. Mayo Clin Proc.
have differential responses to Na+ channel blockade and to 2001;76:299-309.
increases in heart rate: implications for gene-specific therapy. 42. Hess P, Cooper D. Impact of pharmacogenomics on the
Circulation. 1995;92:3381-3386. clinical laboratory. Mol Diagn. 1999;4:289-298.

34 Am J Clin Pathol 2003;119:26-36 © American Society for Clinical Pathology


34 DOI: 10.1092/VMLL66Y5KHQ35KUE
Basic Science / REVIEW ARTICLE

43. Harkin DP. Uncovering functionally relevant signaling 65. Burchiel SW, Knall CM, Davis JW II, et al. Analysis of
pathways using microarray based expression profiling. genetic and epigenetic mechanisms of toxicity: potential roles
Oncologist. 2000;5:501-507. of toxicogenomics and proteomics in toxicology. Toxicol Sci.
44. Snijders AM, Meijer GA, Brakenhoff RH, et al. Microarray 2001;59:193-195.
techniques in pathology: tool or toy? Mol Pathol. 2000;53:289- 66. Hamadeh HK, Amin RP, Paules RS, et al. An overview of
294. toxicogenomics. Curr Issues Mol Biol. 2002;4:45-56.
45. Raetz EA, Moos PJ, Szabo A, et al. Gene expression profiling: 67. Mitcheson JS, Chen J, Lin M, et al. A structural basis for
methods and clinical applications in oncology. Hematol Oncol drug-induced long QT syndrome. Proc Natl Acad Sci U S A.
Clin North Am. 2001;15:911-930. 2000;97:12329-12333.
46. Polyak K, Riggins GJ. Gene discovery using the serial analysis 68. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of
of gene expression technique: implications for cancer chemotherapy plus a monoclonal antibody against HER2 for
research. J Clin Oncol. 2001;19:2948-2958. metastatic breast cancer that overexpresses HER2. N Engl J
47. Lewis F, Maughan NJ, Smith V, et al. Unlocking the archive Med. 2001;344:783-792.
gene expression in paraffin-embedded tissue. J Pathol. 69. Lohrisch C, Piccart M. HER2/neu as a predictive factor in
2001;195:66-71. breast cancer. Clin Breast Cancer. 2001;2:129-137.
48. Maughan NJ, Lewis FA, Smith V. An introduction to arrays. 70. Druker BJ. STI571 (Gleevec) as a paradigm for cancer
J Pathol. 2001;195:3-6. therapy. Trends Mol Med. 2002;8(4 suppl):S14-S18.
49. Alizadeh AA, Ross DT, Perou CM, et al. Towards a novel 71. Joensuu H. Treatment of inoperable gastrointestinal stromal
classification of human malignancies based on gene tumor (GIST) with imatinib (Glivec, Gleevec). Med Klin.
expression patterns. J Pathol. 2001;195:41-52. 2002;97(suppl 1):28-30.

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016


50. Ramaswamy S, Golub TR. DNA microarrays in clinical 72. Baselga J. Targeting the epidermal growth factor receptor with
oncology. J Clin Oncol. 2002;20:1932-1941. tyrosine kinase inhibitors: small molecules, big hopes
51. Schena M, Shalon D, Davis RW, et al. Quantitative [editorial]. J Clin Oncol. 2002;20:2217-2219.
monitoring of gene expression patterns with a complementary 73. Fox SB, Gasparini G, Harris AL. Angiogenesis: pathological,
DNA microarray. Science. 1995;270:467-470. prognostic, and growth-factor pathways and their link to trial
52. Zanders ED. Gene expression analysis as an aid to the identifi- design and anticancer drugs. Lancet Oncol. 2001;2:278-289.
cation of drug targets. Pharmacogenomics. 2000;1:375-384. 74. Herbst RS, Langer CJ. Epidermal growth factor receptors as a
53. Weinstein JN. Pharmacogenomics: teaching old drugs new target for cancer treatment: the emerging role of IMC-C225
tricks [editorial]. N Engl J Med. 2000;343:1408-1409. in the treatment of lung and head and neck cancers. Semin
54. Chicurel ME, Dalma-Weiszhausz DD. Microarrays in Oncol. 2002;29:27-36.
pharmacogenomics: advances and future promise. 75. McDevitt MR, Barendswaard E, Ma D, et al. An alpha-
Pharmacogenomics. 2002;3:589-601. particle emitting antibody ([213Bi]J591) for
55. Los G, Yang F, Samimi G, et al. Using mRNA expression radioimmunotherapy of prostate cancer. Cancer Res.
profiling to determine anticancer drug efficacy. Cytometry. 2000;60:6095-6100.
2002;47:66-71. 76. Sievers EL, Linenberger M. Mylotarg: antibody-targeted chemo-
56. Slonim DK. Transcriptional profiling in cancer: the path to therapy comes of age. Curr Opin Oncol. 2001;13:522-527.
clinical pharmacogenomics. Pharmacogenomics. 2001;2:123-136. 77. Piecyk M, Anderson P. Signal transduction in rheumatoid
57. Dougherty ER, Barrera J, Brun M, et al. Inference from arthritis. Best Pract Res Clin Rheumatol. 2001;15:789-803.
clustering with application to gene-expression microarrays. 78. Williamson AA, McColl GJ. Early rheumatoid arthritis: can
J Comput Biol. 2002;9:105-126. we predict its outcome? Intern Med J. 2001;31:168-180.
58. Fryer RM, Randall J, Yoshida T, et al. Global analysis of gene 79. Watts DA, Satsangi J. The genetic jigsaw of inflammatory
expression: methods, interpretation, and pitfalls. Exp Nephrol. bowel disease. Gut. 2002;50(suppl 3):III31-III36.
2002;10:64-74. 80. Satsangi J. Genetics of inflammatory bowel disease: from
59. Lightcap ES, McCormack TA, Pien CS, et al. Proteasome bench to bedside? Acta Odontol Scand. 2001;59:187-192.
inhibition measurements: clinical application. Clin Chem.
81. Patel VB, Robbins MA, Topol EJ. C-reactive protein: a
2000;46:673-683.
“golden marker” for inflammation and coronary artery disease.
60. Adams J. Development of the proteasome inhibitor PS-341. Cleve Clin J Med. 2001;68:521-524.
Oncologist. 2002;7:9-16.
82. Feinstein SB, Voci P, Pizzuto F. Noninvasive surrogate markers
61. Elliott PJ, Ross JS. The proteasome: a new target for novel of atherosclerosis. Am J Cardiol. 2002;89:31C-43C.
drug therapies. Am J Clin Pathol. 2001;116:637-646.
83. Stein E. Laboratory surrogates for anti-atherosclerotic drug
62. Albanell J, Rojo F, Averbuch S, et al. Pharmacodynamic development. Am J Cardiol. 2001;87:21A-26A.
studies of the epidermal growth factor receptor inhibitor
ZD1839 in skin from cancer patients: histopathologic and 84. McCarthy MI, Hattersley AT. Molecular diagnostics in
molecular consequences of receptor inhibition. J Clin Oncol. monogenic and multifactorial forms of type 2 diabetes. Expert
2002;20:110-124. Rev Mol Diagn. 2001;1:403-412.
63. Hamadeh HK, Bushel P, Paules R, et al. Discovery in 85. Berger A, Preiser W. Viral genome quantification as a tool for
toxicology: mediation by gene expression array technology. improving patient management: the example of HIV, HBV,
J Biochem Mol Toxicol. 2001;15:231-242. HCV and CMV. J Antimicrob Chemother. 2002;49:713-721.
64. Fielden MR, Zacharewski TR. Challenges and limitations of 86. Vandamme AM, Houyez F, Banhegyi D, et al. Laboratory
gene expression profiling in mechanistic and predictive guidelines for the practical use of HIV drug resistance tests in
toxicology. Toxicol Sci. 2001;60:6-10. patient follow-up. Antivir Ther. 2001;6:21-39.

© American Society for Clinical Pathology Am J Clin Pathol 2003;119:26-36 35


3 DOI: 10.1092/VMLL66Y5KHQ35KUE 35
Ross and Ginsburg / THE INTEGRATION OF MOLECULAR DIAGNOSTICS WITH THERAPEUTICS

87. Buss N, Cammack N. Measuring the effectiveness of 94. Bernard PS, Wittwer CT. Real-time PCR technology for
antiretroviral agents. Antivir Ther. 2001;6:1-7. cancer diagnostics. Clin Chem. 2002;48:1178-1185.
88. Russo A, Zanna I, Tubiolo C, et al. Hereditary common 95. Jain KK. Applications of proteomics in oncology.
cancers: molecular and clinical genetics. Anticancer Res. Pharmacogenomics. 2000;1:385-393.
2000;20:4841-4851. 96. Lee KH. Proteomics: a technology-driven and technology-
89. von Knebel Doeberitz M. New molecular tools for efficient limited discovery science. Trends Biotechnol. 2001;19:217-222.
screening of cervical cancer. Dis Markers. 2001;17:123-128. 97. Herrmann PC, Liotta LA, Petricoin EF III. Cancer
90. Ross JS. Economic, regulatory, and practice issues in proteomics: the state of the art. Dis Markers. 2001;17:49-57.
molecular pathology and diagnostics. Am J Clin Pathol. 98. Srinivas PR, Srivastava S, Hanash S, et al. Proteomics in early
1999;112(1 suppl 1):S7-S10. detection of cancer. Clin Chem. 2001;47:1901-1911.
91. Sakallah SA. Molecular diagnostics of infectious diseases: 99. Petricoin EF, Ardekani AM, Hitt BA, et al. Use of proteomic
state of the technology. Biotechnol Annu Rev. 2000;6:141-161. patterns in serum to identify ovarian cancer. Lancet.
92. Pang CP. Molecular diagnostics for cardiovascular disease. 2002;359:572-577.
Clin Chem Lab Med. 1998;36:605-614. 100. Phelps ME. PET: the merging of biology and imaging into
93. Lacroix J, Doeberitz MK. Technical aspects of minimal molecular imaging. J Nucl Med. 2000;41:661-681.
residual disease detection in carcinoma patients. Semin Surg
Oncol. 2001;20:252-264.

Downloaded from http://ajcp.oxfordjournals.org/ by guest on July 5, 2016

36 Am J Clin Pathol 2003;119:26-36 © American Society for Clinical Pathology


36 DOI: 10.1092/VMLL66Y5KHQ35KUE

You might also like