18.12.2012 Views

2012 EDUCATIONAL BOOK - American Society of Clinical Oncology

2012 EDUCATIONAL BOOK - American Society of Clinical Oncology

2012 EDUCATIONAL BOOK - American Society of Clinical Oncology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Why Hasn’t Genomic Testing Changed the<br />

Landscape in <strong>Clinical</strong> <strong>Oncology</strong>?<br />

By Daniel F. Hayes, MD, Muin J. Khoury, MD, PhD, and David Ransoh<strong>of</strong>f, MD<br />

Overview: The “omics” revolution produced great optimism<br />

that tumor biomarker tests based on high-order analysis <strong>of</strong><br />

multiple (sometimes thousands) <strong>of</strong> factors would result in truly<br />

personalized oncologic care. Unfortunately, 10 years into the<br />

revolution, the promise <strong>of</strong> omics-based research has not yet<br />

been realized. The factors behind the slow progress in omicsbased<br />

clinical care are many. First, over the last 15 years,<br />

there has been a gradual recognition <strong>of</strong> the importance <strong>of</strong><br />

conducting tumor biomarker science with the kind <strong>of</strong> rigor that<br />

has traditionally been used for therapeutic research. However,<br />

this recognition has only recently been applied widely, and<br />

therefore most tumor biomarkers have insufficiently high<br />

levels <strong>of</strong> evidence to determine clinical utility. Second, omicsbased<br />

research <strong>of</strong>fers its own particular set <strong>of</strong> concerns,<br />

TUMOR BIOMARKERS are used to determine a patient’s<br />

current status and more importantly to predate<br />

future events that might be modified by intervention. 1<br />

Tumor biomarkers can be analyzed in cancer or healthy<br />

tissue, in secretions, and circulating in blood. Tumor biomarkers<br />

usually represent somatic changes that have<br />

emerged during the process <strong>of</strong> carcinogenesis. However, it is<br />

also reasonable to consider inherited germ-line differences<br />

between individuals that predict higher risk <strong>of</strong> developing a<br />

new malignancy or for estimating differential distribution,<br />

metabolism, or response to a drug (“pharmacogenomics”). 2,3<br />

Assays for tumor biomarkers can identify changes, or<br />

individual differences, in nucleic acids (DNA, RNA), proteins,<br />

lipids, whole cells, or tissue processes. Until recently,<br />

an assay for a biomarker usually analyzed a single analyte,<br />

or substance. Perhaps one <strong>of</strong> the best examples is the<br />

development <strong>of</strong> assays for the estrogen receptor (ER) to<br />

predict both prognosis and likelihood <strong>of</strong> responding to antiestrogen,<br />

or “endocrine” therapies. 4,5 The biology <strong>of</strong> ER was<br />

determined in the 1960s, and the first assay was a cumbersome<br />

test to biochemically measure binding <strong>of</strong> radioactively<br />

labeled estrogen to the receptor. Subsequent tests using<br />

specific antibodies to perform either enzyme-linked immunosorbent<br />

assays (ELISAs) and more recently immunohistochemistry<br />

(IHC) replaced the original ligand-binding<br />

assays, and are now almost uniformly used in clinical<br />

medicine. However, recently, newer assays that measure<br />

RNA expression have been introduced. Regardless, these<br />

are all tests that assay for a single analyte, ER, and not for<br />

several analytes that might be combined into a unified index<br />

designed to make a clinical decision.<br />

High-Dimensional Biomarkers<br />

In addition to measuring a single analyte, assessment <strong>of</strong><br />

complex processes, such as counting vessels to determine<br />

levels <strong>of</strong> angiogenesis, or development <strong>of</strong> a multifactoral<br />

index, such as tumor grade (which combines estimates <strong>of</strong><br />

relative gland formation, nuclear appearance, and mitotic<br />

rate), represent higher-order forms <strong>of</strong> a single test. In this<br />

regard, during the last decade, advances in molecular biology,<br />

technology, and bioinformatics have led to a new field<br />

loosely described as “omics.” This field encompasses several<br />

disciplines, generating high-dimensional data from global<br />

e52<br />

especially in regard to overfitting computational models and<br />

false discovery rates. Researchers and clinicians need to<br />

understand the importance <strong>of</strong> analytic validity, and the difference<br />

between clinical/biologic validity and clinical utility. The<br />

latter is required to introduce a tumor biomarker test <strong>of</strong> any<br />

kind (single analyte or omics-based), and are ideally generated<br />

by carefully planned and properly conducted “prospective<br />

retrospective” or truly prospective clinical trials. Only<br />

carefully planned studies, which take all three <strong>of</strong> these into<br />

account and in which the investigators are aware and recognize<br />

the enormous risk <strong>of</strong> unintended bias and overfitting<br />

inherent in omics-based test development, will ultimately<br />

result in translation <strong>of</strong> the exciting new technologies into<br />

better care for patients with cancer.<br />

sets <strong>of</strong> biologic molecules such as DNAs (“genomics”), RNAs<br />

(“transcriptomics”), proteins (“proteomics”), and metabolites<br />

(“metabolomics”). 6 Massive amounts <strong>of</strong> data are used to<br />

produce a pr<strong>of</strong>ile, or “signature,” generated by a computational<br />

mathematical function model. This model may be<br />

unsupervised, meaning that specimens are grouped computationally<br />

by apparent similarities in the omics patterns,<br />

without regard to preconceived biologic or clinical associations.<br />

Alternatively, generation <strong>of</strong> the pr<strong>of</strong>iles can be “supervised”;<br />

in this case, the signature is “pegged” to some sort <strong>of</strong><br />

prospectively defined biologic or clinical characteristic <strong>of</strong><br />

interest. Ultimately, at least in regard to clinical care, one or<br />

more omics-based test that reputedly has clinical utility in<br />

guiding patient care is generated.<br />

In an online continuous horizon scanning review <strong>of</strong> the<br />

literature from 2009 to the present, researchers from the<br />

Centers for Disease Control and Prevention found more than<br />

400 new genomic and other omics-based tests in transition<br />

from bench to bedside, <strong>of</strong> which the vast majority are related<br />

to cancer. 7 However, in <strong>2012</strong>, few if any omics-based tests<br />

have actually been widely adopted or embraced in the clinic.<br />

Why not? There are several obstacles that block introduction<br />

and use <strong>of</strong> an omics-based test. These relate to generation<br />

and validation <strong>of</strong> tumor biomarker tests in general, but<br />

in addition omics-based tests have special considerations<br />

that have impeded progress in the field. 8 It is essential that<br />

basic, translational, clinical, and computational scientists,<br />

and importantly clinicians caring for patients with cancer,<br />

understand these obstacles and work to overcome them so<br />

that patients receive better, more personalized oncologic<br />

care than they do now.<br />

From the University <strong>of</strong> Michigan Comprehensive Cancer Center, Ann Arbor, MI; Epidemiology<br />

and Genomics Research Program, Division <strong>of</strong> Cancer Control and Population<br />

Sciences, National Cancer Institute, Bethesda, MD; Office <strong>of</strong> Public Health Genomics,<br />

Centers for Disease Control and Prevention, Atlanta, GA; Departments <strong>of</strong> Medicine and<br />

Epidemiology, University <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC.<br />

Authors’ disclosures <strong>of</strong> potential conflicts <strong>of</strong> interest are found at the end <strong>of</strong> this article.<br />

Address reprint requests to Daniel F. Hayes, MD, Breast <strong>Oncology</strong> Program, University<br />

<strong>of</strong> Michigan Comprehensive Cancer Center, 6312 Cancer Center, 1500 E. Medical Center<br />

Drive, Ann Arbor, MI 48109-0942; email: hayesdf@umich.edu.<br />

© <strong>2012</strong> by <strong>American</strong> <strong>Society</strong> <strong>of</strong> <strong>Clinical</strong> <strong>Oncology</strong>.<br />

1092-9118/10/1-10

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!