Vous êtes sur la page 1sur 3

BSR|ProfMa.

Biomarker Technology
Luisa G. Daroy || August 11, 2017
Block #1

o Clinical cancer therapeutics led to the use of lab tests to


TOPIC OUTLINE distinguish responders from non-responders to targeted
I. History therapies
A. Early biomarkers o Ex. Philadelphia chromosome in imatinib response in
B. Era of Molecular Biology CML(Chronic Myelogenous Leukemia) cases
C. 1980s 1990s: Molecular Biomarkers o Ex. Her2/neu tyrosine kinase receptor for Herceptin
D. Scientific &Technological Advances in Biomarker response in breast cancer patients
Research
E. Biomarkers Consortium in 2006 D. Scientific&Technological Advances in Biomarker
II. Definition of Terms Research
III. Biomarker Development Pathway Completion of the Human Genome Project provided
A. Pathway reference data of human DNA sequences and of other
B. Advantages of Biomarker Development organisms from which were derived certain biomarkers of
C. Genetic Biomarkers disease.
D. Protein Biomarkers
E. Proteome and Proteomics
E. Biomarkers Consortium (2006)
IV. Bioinformatics
V. References Public-private initiative with industry and government to spur
VI. Quiz biomarkers development and validation projects in cancer,
PPT Audio Book CNS, and metabolic disorders.

TRANSERS NOTE: The part IV Bioinformatics of this trans is II. Definition of Terms
in book trans format since it was skipped by the lecturer. A. Biomarker
A biologic indicator of health or disease.
I. HISTORY OF BIOMARKERS A characteristic that is measured and evaluated objectively
Term introduced by Karpetsky, Humphrey and Levy in April as an indicator of normal biologic processes, pathogenic
1977 edition of J National Cancer Institute processes, or pharmacologic response to therapeutic
7th C, B.C.: Diabetes first associated with sweetness of intervention.
urine
B. Surrogate Endpoint Marker
A. Early Biomarkers A biomarker that can substitute for a clinical endpoint and is
Uroscopy expected to predict clinical benefit or harm, or lack of benefit
Blood pressure or harm, based on epidemiologic, therapeutic,
Imaging pathophysiologic, or scientific evidence.
Electrocardiography (2020) something you can observe clinically such as
Hematology cardiac troponins, CK-MB for myocardial infarction
Blood and urine chemistry
C. Uses of Biomarkers
B. Era of Molecular Biology Diagnosing, classifying or grading the severity of disease in
26 February 1953, Cambridge Watson and Crick model of both clinical and laboratory settings
DNA structure Provide efficacy, toxicity, and mechanistic info for the
o 1972: 1st gene sequence (of a bacterium) preclinical and clinical phases of drug discovery
o 1976: 1st complete RNA genome sequence Together with therapeutics, may be applied to produce
o 1977: 1st complete DNA genome sequence commercial tests that aid patient selection or drug dosing
Genomics, Proteomics, Metabolomics foundations for (personalized medicine)
the molecular basis of disease
With the advent of molecular technology, the discovery of D. Biomarker Tests
DNA molecule in 1953 by Watson and Crick spawned the The best biomarkers are:
revolution in biology and medicine in which you have o Accurate
complete sequences of the genes which encode for life o Relatively non-invasive
processes coming one after the other, laid the foundations in o Easy-to-perform tests that can be done at the bedside or
genomics, proteomics, metabolomics for the molecular basis in an outpatient setting
of the disease. Examples:
Major Scientific Contributions and Research o Blood or spot urine specimens that can be measured
Infrastructure Supporting Biomarker Discovery: These serially and have a fast turnaround
are the technologies that gave rise to new novel biomarkers o (2020) rapid PCR test instead of TB culture
as we now analyze it the medical practice
o Human Genome Project E. Biomarkers
o Mouse models of disease (recombinant DNA technology)
o Information management (informatics tools, open-source Any parameter of a patient that can be measured:
databases, open-source publishing, biomarker reference o Genetic mutations
services. o Gene polymorphisms
o Population-based studies and gene-environment o mRNA expression profiles
interaction studies o Epigenetic markers
o Computational biology and biophysics o Imaging methods
o Medical imaging: structural and functional o Electrical signals
o High-throughput technologies - where you have massive o Lipids
volumes of data being produced in vitro cell-based o Proteomic patterns
screening, nanotechnology platforms, molecular o Proteins
separation techniques, robotics, automated microassays,
high-resolution optics F. Requirements for Biomarker Research
o Proteomics, metabolomics, epigenomics Innovations in technology applications
o Pharmacogenomics New statistical methods and clinical research designs
o Molecular toxicology Data management and informatics
o Genome-wide association studies Clinical registries
o Molecular pathways systems biology and systems Repositories of biological specimens
engineering Imaging files
Common reagents
C. 1980s-1990s: Molecular Biomarkers
Viral and Immune Indicators of disease G. Requirements for Successful Biomarker Development
o CD4+ T-lymphocyte counts in HIV cases were used as Access to sufficient, well-characterized sample biobanks
surrogate endpoints (repositories of sufficient and well-characterized samples of
Target-designed therapies biological specimens)
o Companion diagnostics for cancer therapeutics Understanding of the entire complex biomarker development
process.

Trans #2 Barredo | Del Mundo|| Checked by: Fernan


Page 1 of 3
BSR Biomarker Technology
Use of multi-disciplinary team approach.
Validation of every step of both assay performance and B. Advantages of Biomarker Development
diagnostic utility. Compared with the cost and risk of drug development,
biomarkers offer the opportunity to have an impact on patient
III. BIOMARKER DEVELOPMENT PATHWAY health in a more economical manner and may provide an
A. PATHWAY opportunity to speed up the drug development process.
Step 1. Understand and Define the Disease Biomarker development leads to earlier disease detection
Detailed knowledge of the disease: definition, differential and prediction of which patients will respond to which
diagnosis, disease subsets, and the local or systemic therapies.
responses.
Reliable case definition is very valuable: may be extremely C. Genetic Biomarkers
specific or based on a constellation of symptoms and signs Genetic variation gives rise to unique DNA profiles of each
(systems-based), should include a temporal element. individual
If the disease is poorly defined, then the newly discovered o Mutations
biomarker may ultimately change the definition of the o Single nucleotide polymorphisms (SNPs)
disease. o Copy number variants (CNV)
o Microsatellites
Step 2. Frame the question: What critical information will o mRNA expression profiles
the biomarker provide? o Epigenetic markers, i.e. DNA methylation patterns
The best biomarkers serve a basic science, translation or o Micro RNA (miRNA)
clinical need that advances the diagnosis, prevention or o Circulating tumor cells
treatment of the disease.
A biomarker may be targeted at early detection of disease or D. Protein Biomarkers
to monitor the stage, severity, progression or regression of Protein alterations in disease may occur in many different
disease after diagnosis. ways that are not predictable from genomic analysis; a better
A biomarker may be targeted to predict drug response or understanding of these alterations in a protein level will have
follow the effect of an intervention. (the best question may substantial impact in medicine
not be the most obvious question). Benefits
o Better understanding of disease process
Step 3. Desired site of clinical measurement o Development of new biomarkers for diagnosis and early
Biomarkers can be assayed in easily obtainable fluids; detection of disease
serum, plasma or urine, or other sites, such as saliva, sweat, o Accelerated development of new drugs
hair and biopsy material. Sometimes a little bit more
invasive: aspirates, CSF, synovial fluid, bone marrow.
E. Proteome and Proteomics
Choice is driven by a balance between clinical relevance,
ease of collection and stability versus disease specificity and
analytical simplicity of the discovery step.

Step 4. Devise a strategy for the discovery process


The biomarker discovery strategy is influenced by the
diagnostic difficulty, disease variability, subclasses,
biomarker goals, ultimate site of measurement, and
discovery platform.
Questions that you have to answer:
o Should the discovery process start on the diseased tissue
or the material that will make up the final clinical assay?
So for example in cancer, should it start in a tumor, which
is highly invasive to get or should it start with the blood?
Should human samples or samples from animal models
be used?
o Should subtractive comparison between normal and
diseased or before and after samples be used, or should
comparison of several groups of samples be made in the
discovery step?

Step 5. Which samples should be used for the discovery Figure #1. Steps in Protein Analysis
phase?
1. PROTEOME
Samples should match the clinical question as closely as
possible. The entire protein complement in a given cell, tissue or
organism.
Samples should mirror the disease process being
2. PROTEOMICS
investigated.
A global approach to the molecular biology study of the
Samples should come from carefully defined sources, be of
overall distribution of proteins in cells, identification and
high quality, sufficient in quantity and carefully preserved.
characterization of individual proteins of interest, and the
Step 6. Determine which discovery method to use elucidation of their relationships and functional roles
(example: biomarkers for heart disease: apolipoprotein,
Measurement of mRNA expression.
cytokine protein, interleukin, cystatin C).
2D differential in-gel electrophoresis (proteins)
Characterize the information flow through the protein
Surface-enhanced laser desorption ionization (SELDI) mass circuitry that interconnects the extracellular
spectrometry. microenvironment to the serum or plasma macro-
Liquid Chromatography-Tandem mass spectrometry environment thru intracellular signaling systems and their
Tissue microarray control of gene transcription.
Single nucleotide polymorphisms (SNPs) detection Functional Proteomics function of a particular protein
Differential Proteomics differentiates between diseased
Step 7. Review the significance and feasibility and normal or before and after
By studying global patterns of protein content and activity,
Step 8. Perform the experiments and prioritize the hit list
and how these change during development or in response
to disease, proteomics research is poised to boost our
Step 9. Develop a robust clinical assay and initial clinical
understanding of systems-level cellular behavior.
evaluation to detect existing disease
Clinical research also hopes to benefit from proteomics by
both the identification of new drug targets and the
Step 10. Evaluate the clinical utility
development of new diagnostic markers.
3. GOALS OF PROTEOMICS
Step 11. Combine biomarker with clinical data and other
biomarkers. To define the identities, quantities, structures and
functions of complete complements of proteins.
To characterize how these properties vary in different
cellular contexts.

Trans #2 Barredo | Del Mundo|| Checked by: Fernan Page 2 of 3


BSR Biomarker Technology
Such studies enable genetic, biochemical and cell Reproducibility and validation of tumor biomarkers should
biological technologies to be applied on a systemic level, be addressed carefully.
leading to the assignment of biochemical activities, the Standardization of sampling and analytical techniques
construction of protein arrays, the identification of
interactions, and the localization of proteins within cellular
Integration of proteomic, genomic, and metabolomics-level
compartments. data.
Cost: R&D expenditure was at $5.3B (2009).
IV. BIOINFORMATICS Need to develop multiplex assay systems.
All in all, biomarkers will be composed of these different
signals and integrated data base. Medicine is becoming E. Biomarkers in the future
evidence-based and at a molecular level.
Understanding the molecular basis of tumor characteristics
such as neoplasia development, metastases, invasion and
A. Applications resistance to therapy will play an important role in the
PROFILING OF DISEASE TISSUES development of personalized cancer therapy.
o Classification of leukemia into different subtypes New biomarker technologies combined with advanced
o Changes in myocardial proteins associated with heart bioinformatics is being used to identify molecular signatures
failure in humans and anima l models (rat, monkeys) of individual tumors based on protein pathways and
SERUM BIOMARKERS signaling cascades.
o Detection of low-abundance and potentially new The discovery of new highly sensitive and specific
circulating disease marker proteins biomarkers for early disease detection and risk stratification
DIFFERENTIAL IN-GEL ELECTROPHORESIS coupled with personalized therapies holds the key to future
(DIGE) cancer treatment.
o Quantification of protein expression between esophageal V. REFERENCES
CA cells and normal epithelial cells Lecturers PPT
DISEASE BIOMARKER IDENTIFICATION 2020 Trans
o Comparative analysis of protein expression in normal VI. QUIZ
anddisease tissues to identify aberrantly expressed 1. The following are the foundations for the molecular basis of
proteins thatmay represent new markers disease except:
o Analysis of secreted proteins in cell lines and A. Genomics
primarycultures B. Metabolomics
o Direct serum protein profiling by mass spectrometry C. Epigenomics
(ex.Auto-antibodies against tumor proteins) D. Proteomics
o Identificationoftumorantigens 2. Which of the following is the 5th step of biomarker
development pathway?
BIOMARKER VALIDATION A. Review the significance and feasibility
o Prospective, well controlled clinical studies of B. Develop a robust clinical assay and initial clinical
diversepatients across multiple institutions, with well- evaluation to detect existing disease
established standards for all steps in the process C. Understand and Define the Disease
Tissue collection D. Determine which samples should be used for the
Purification discovery phase
Amplification 3. Which of the following is not a requirement for a successful
Hybridization or ligand-binding biomarker development?
Data capture A. Repositories of biological specimens
B. Understanding of the entire complex biomarker
Normalization development process.
Statistical analysis C. Use of multi-disciplinary team approach.
Scoring D. Validation of every step of both assay performance and
o Reproducibility within and among laboratories diagnostic utility.
4. Which biomarker is considered a companion diagnostics for
B. Chronology of Biomarker Development cancer therapeutics?
A. Target-designed therapies
A biomarker is first identified then evaluated for a particular B. Single nucleotide polymorphisms (SNPs)
clinical indication. C. CD4+ T-lymphocyte
Analytical and clinical validations must be performed D. DNA methylation patterns
before submission for US FDA approval. 5. What describes a proteome?
Alternatively, the marker might bypass the FDA approval A. A collection of cells used for genetic tests
process if it is to be used for research purposes only. B. A biologic indicator of health or disease
C. Protein alterations in disease
Once approved, the Centre for Medicaid and Medicare D. The entire protein complement in a given cell, tissue or
Services (CMS) might determine that it is reasonable and organism
necessary for improved patient care and therefore,
reimbursable.
Because CMS decisions indirectly influence coverage by
private insurance carriers, a marker is not widely used in the Answers: 1.C, 2.D, 3.A. 4.B, 5. D
clinic unless all of the steps in the process have been
completed.

C. Regulatory Bodies
FDA: in vitro diagnostics (IVD) safety and effectiveness
of an IVD refers to the consequences expected from reliance
on it to make clinically significant diagnostic or treatment
decisions. i.e. tumor markers.
Center for Medicaid and Medicare Services: Analyte-
specific reagents (ASRs) biomarkers used in in-house
clinical laboratory for restricted for research only purposes

D. Challenges
Biological variability among patient samples as well as the
huge dynamic range of biomarker concentrations is the main
challenge to deduce diagnostic patterns unique to specific
cancer states.

Trans #2 Barredo | Del Mundo|| Checked by: Fernan Page 3 of 3

Vous aimerez peut-être aussi