Introduction To Molecular Pathology


Key Points

  • The revolution in molecular biology continues to have a profound impact on the field of anatomic and clinical pathology (diagnostic medicine).

  • Techniques, including polymerase chain reaction and other amplification and hybridization methods, are capable of detecting DNA in very small quantities in tissue and body fluids, enabling detection of various disease states, including cancer.

  • The human genome project has created robust high-throughput and highly sensitive genetic methods for detecting abnormal genes in patient specimens and for establishing patterns of gene expression that are characteristic of specific diseases.

  • Proteomic methods detecting protein expression in serum samples from patients allow for diagnosis of different types of cancers.

  • Gene array high-throughput sequencing and proteomic methodologies require sophisticated mathematical methods of pattern recognition that allow for detection of disease.

Introduction

During the past decade, a vast amount of knowledge has been gained regarding genes, gene products, and their role in human disease. This knowledge has allowed us to better understand many disease processes and to start defining diseases and disease processes in terms of their molecular pathogenesis. Molecular pathology refers to the analysis of nucleic acids and proteins to diagnose disease, predict the occurrence of disease, predict the prognosis of diagnosed disease, and guide therapy. Molecular pathology–based diagnostics testing is a relatively recent specialty of laboratory medicine and is still in a state of flux at many levels, such as the choice of test, technology, automation, and reimbursement. The scope of molecular testing has expanded rapidly since its introduction in the late 1980s. This has led to the development of new clinical molecular assays for use in diagnosis, prognosis, selection of therapeutic modalities, and monitoring of disease in both general and personalized fashions ( , ). The technologies that constitute molecular diagnostics—such as first-generation amplification, next-generation DNA sequencing, DNA probes, fluorescence in situ hybridization (FISH), second-generation biochips and microfluidics, next-generation signal detection, biosensors, and molecular labels—are influencing the discovery of therapeutic molecules, the screening and diagnosis of patients, and the optimization of drug therapy. In the past few years, this rapidly evolving field has seen several fascinating developments. There are many applications of this technology to different areas of the clinical laboratory. Whereas testing for inherited genetic diseases, cancer, viral load, and infectious diseases continues to predominate, the impact of pharmacogenomics on molecular diagnostics continues to evolve and impact diagnostics and prognostics of disease. The operation of a clinical molecular pathology laboratory requires integration of expertise in medical, scientific, and clinical molecular pathology; resources, including facilities, equipment, and personnel; and skills in organization, administration, management, and communication.

Special Considerations For Molecular Diagnostics Laboratories

Whereas all molecular diagnostic tests share the use of nucleic acid technology, the different clinical applications may require different management considerations.

Infectious Disease

Increasing numbers of microorganisms are detected or characterized by molecular methods, including mass spectroscopy, as discussed in Part 7, which in many cases have become the standard of practice, especially for microorganisms that are difficult to culture or when subsequent culture is needed to identify drug resistance ( ). As the volume of testing has grown, a need has arisen for automation of different steps of testing and standardized kits. There has been a surge of different automation platforms. Automated platforms perform nucleic acid extraction from different types of specimens, automated liquid handling systems set up reactions, and real-time technology monitors and quantifies nucleic acids as they are being amplified. In addition, quantitative molecular methods based on polymerase chain reaction (PCR) amplification and automated rapid sequencing techniques that are essential for viral speciation have become the standard of practice in monitoring response to therapy, especially in patients with human immunodeficiency virus (HIV), hepatitis B virus (HBV), and hepatitis C virus (HCV) infections. With these automated methodologies, the need for confidentiality, especially for patients undergoing HIV testing, has prompted specific federal and state regulations to ensure the protection of patients and their families. In addition, each state might have a list of different organisms that, if detected, must be reported to the state health department.

Cancer

Molecular methods are currently used for hematopoietic neoplasms as well as for solid tumors. The nucleic acid changes, either of DNA or ribonucleic acid (RNA), are present only in the affected populations of cells and are not part of the genetic makeup of the individual. Current technologies—such as real-time PCR, next-generation DNA sequencing, and microarray analysis—have provided greater diagnostic sensitivity and specificity for diagnostic testing, prognosis, treatment selection, monitoring response to therapy, and detection of minimal residual disease. In addition, the exquisite sensitivity of these molecular methods has allowed the testing of very small amounts of nucleic acid from a wide range of sample types. Formalin-fixed paraffin-embedded tissue can provide good quality and quantity of DNA and RNA from processed tissues. However, very specific protocols either for DNA or RNA extraction must be followed. In contrast to inherited genetic testing that needs to be done once per lifetime, molecular oncology tests often are performed repeatedly (e.g., for initial diagnosis and during and after treatment for monitoring response to therapy). Molecular methods that identify rearrangements or mutations that cause specific tumor types may also be used to assess minimal residual disease. Molecular test results should be interpreted in the context of other laboratory testing, such as histopathology, flow cytometry, and clinical findings. The detection of a specific mutation in the tumor cells may also be used to determine the appropriate course of therapy. This is the case for metastatic colon cancer. A number of studies have shown that anti–epidermal growth factor receptor (anti-EGFR) monoclonal antibodies are effective treatments for metastatic colorectal cancer, but only in patients with the wild-type oncogene KRAS . The anti-EGFR monoclonal antibodies panitumumab and cetuximab are approved in the United States for treatment of metastatic colorectal cancer refractory to chemotherapy but are not recommended for use in patients with mutations in KRAS codon 12 or 13. Similarly, panitumumab is approved for the treatment of metastatic colorectal cancer only in patients with wild-type KRAS in Europe and Canada. It is clear that KRAS mutational analysis has become an important aspect of disease management in patients with metastatic colorectal cancer ( ; ). Laboratory issues specific for hematopoietic neoplasms and solid tumor testing include the need for fresh or frozen tissue for RNA-based testing, the use of microdissection to reduce nonmalignant cell population in the specimens, and the need to work with small tissue specimens, such as needle biopsies.

Inherited Disorders

All diseases have a genetic contribution, whether it is a specific genetic disease or an increased likelihood for developing a medical condition. Genetic disorders are primarily the result of a germline mutation or a mutation present in every cell of an individual. For this reason, genetic testing has far-reaching implications, because it could affect family members that might have inherited the same mutation. Molecular genetic testing is currently used for diagnosis, carrier status evaluation, and prenatal and presymptomatic DNA testing. Diagnostic testing is usually performed on affected individuals for establishing or confirming a clinical diagnosis.

Carrier testing is performed in an asymptomatic healthy individual to identify whether the individual is a carrier for an autosomal or X-linked recessive condition and whether the individual is at risk for having an affected child. This application can be used for individuals with a family history of a genetic disorder or for population screening, such as in screening for cystic fibrosis (CF). Carrier testing can be done first by testing an affected family member to identify the specific mutation present in the family. Once a specific mutation is identified, family members can be tested for that particular mutation, thus improving the accuracy of the risk assessment for the individuals in that family with a negative test result. In contrast, population screening focuses on the most common or prevalent mutation, most of the time with different rates of detection for different ethnic populations. Prenatal testing is performed to identify a fetus with a genetic disease or condition. Prenatal testing analyzes fetal cells obtained by amniocentesis or by chorionic villus sampling. Genomic technologies such as noninvasive prenatal testing (NIPT) are capable of detecting abnormalities in fetal DNA from maternal blood samples ( ). This type of testing is usually initiated because family history or maternal factors suggest the need for it. In addition, some laboratories offer preimplantation genetic testing in the setting of in vitro fertilization for couples with a family history of a specific genetic disease. Presymptomatic testing is used primarily for the identification of adult onset of a genetic condition that will occur later in life, such as Huntington disease. Presymptomatic testing is the most problematic and challenging type of testing in terms of its psychological effect on the individual. Hence, it requires extensive protocols for pregenetic and postgenetic testing counseling.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here