Genomic Biomarkers: Revolutionizing Diagnosis and Resolution of Periodontal Disease

Special Article – Periodontitis Treatment

J Dent & Oral Disord. 2016; 2(6): 1033.

Genomic Biomarkers: Revolutionizing Diagnosis and Resolution of Periodontal Disease

Luthra S*, Grover HS and Maroo S

Department of Periodontics & Oral Implantology, S.G.T. University, India

*Corresponding author: Shailly Luthra, Department of Periodontics & Oral Implantology, S.G.T. University, Haryana, India

Received: July 21, 2016; Accepted: August 26, 2016; Published: August 29, 2016

Abstract

Periodontal disease encompasses an array of conditions affecting the health of the periodontium. It is one of the most common diseases affecting 10-15% of world’s population. Traditional methods of assessing the periodontal status have been proved to be insufficient in recognizing the prognosis of the disease. Therefore, there is a need for an advanced innovative diagnostic test. Current genomics and biotechnology promise the development of biomarkers to identify individual at higher risk of developing periodontal disease, enable its early detection, and improve diagnostic classification for personalized treatment based on genomic makeup of an individual. It has also been used in the field of pharmacology for monitoring and providing informational feedback for drug discovery and development. This article highlights the types of genomic biomarkers, obstacles to future success and its clinical use towards personalized medicine.

Keywords: Genomic biomarkers; Diagnostic; Periodontitis; Personalized medicine; Pharmacogenomics

Introduction

Periodontal disease is a chronic inflammatory disease of diverse origin that involves degradation of periodontal tissues, including alveolar bone-. It affects 10% to 15% of the total world’s population and can eventually result in tooth loss [1]. Development of periodontal disease requires the presence of bacterial plaque which induces pathological changes in the tissues by both direct (through pathological effects of bacteria and their products on the periodontium) and indirect means (initiation of a number of hostmediated destructive processes. After the initiation of gingival inflammation, the disease progresses with the loss of periodontal ligament and its attachment to cementum leading to the formation of a periodontal pocket, tooth mobility, and subsequent tooth loss if left untreated [2,3].

Conventionally, periodontitis has been treated with numerous therapeutic strategies aimed principally at eradication of periodontal pathogens. These treatment strategies integrate both surgical and non-surgical approaches along with anti-microbial therapy for the management of the periodontal disease. However, according to a study by Haffeeje AD et al. (1997), traditional treatments fail 32% of the time within 9 months [4]. This is firstly because of the inability of the traditional diagnostic tool to assess the actual current disease status3 and secondly because most of the drugs are effective for only 25-60 % of patients [5]. Annual adverse drug reactions in the United States alone are seen in more than 2 million cases, with 100,000 involving deaths [6]. The quest for identifying these variances in the disease morphology and treatment outcome dates back to a century when Garrod first introduced the term “chemical individuality” [7]. Subsequently, many studies were conducted and it was concluded that the differences in the genetic makeup of a population form the basis for most of the disease including periodontitis [8,9]. This hypothesis has led to the development of various pioneering diagnostic tests that focus on the identification of genomic biomarkers so as to identify and quantify the periodontal risk and treatment outcome.

“Genomic Biomarkers - The chronicle of life”

Genomic Biomarkers are the evidence of past or present life as they reflect the entire spectrum of health as well as disease from its earliest manifestations to the terminal stages. For a biomarker to be used as a diagnostic tool, it should have high sensitivity, specificity along with high predictive value [10]. It should be safe and easy to measure accompanied by a verified treatment to modify the biomarker. The follow-up tests should be cost effective as well as consistent across genders and ethnic groups.

Genomic biomarkers comprise tools and technologies that aid in understanding the aetiology, diagnosis, progression, regression, or outcome of treatment of disease.

At present the genomic biomarker arena can be divided into two broad subsets.

The mechanisms by which these biomarkers are elevated in biological fluid include gene overexpression, increased protein secretion and shedding, angiogenesis, invasion and destruction of tissue architecture [32].

Biomarker discovery and omics technology

The medical term “biomarker discovery” describes the process by which biomarkers are identified. Regularly used sources for identifying potential periodontitis biomarkers includes blood, saliva and gingival crevicular fluid.

The recent interest in biomarker discovery is driven by new molecular diagnostic techniques, which have the potential to find appropriate markers rapidly, without detailed insight into the mechanisms of a disease. Genomics, proteomics, transcriptomics, and metabolomics are some technologies used in this process. Secretomics, along with other OMICS tool, also help in high-throughput searches for biomarkers, thus emerging as an important technology. The main advantages of proteomics and other technologies are that low levels of a specific biomarker can be detected, and that this can be achieved easily and noninvasively by using saliva or gingival crevicular fluid [33]. Investigators are designing “lab-on-a-chip” prototypes. This handheld, automated, easy to use integrated system enables simultaneous and rapid detection of multiple salivary protein and nucleic acid targets by using small samples.

Challenges faced in biomarker discovery

Experimental design: It is probably one of the most ignored and least appreciated components of biomarker discovery. Appropriate number of samples should be analysed to achieve statistically significant data outcomes. Adequate controls are a very necessary unit in the design of such studies and can influence, for example, whether a global or targeted analysis is appropriate, and whether tissue or body fluid should be analysed [34].

Sample quality: The quality of samples analysed will ultimately determine the quality of biomarkers produced. A number of factors must be taken into consideration. For example, a clear lineage and adequate care for animals is necessary, whereas in the case of human samples, history, outcomes and storage conditions are all very important. In particular, one must also consider whether to pool samples or analyze individual samples. Though pooling sample makes the experiment very cost effective [35], many researchers believe that pooling is not appropriate as the individual samples provide more important information as they contain intrinsic biological variability [34].

Technology platforms: An old proverb “Garbage in Garbage Out” is popular computing slang for “if the input data is wrong, the results will also be wrong” and a modern modification of this saying “Garbage in = Gospel Out” refers to the blind acceptance of the answer obtained from computers. This can result in faulty decision making and therefore, it is necessary to check and re-check the data and coding to ensure that the results are valid.

a. There has been a tremendous development in -omics platform capability over the past decade. However there still remain a number of concerns. Precision and reproducibility of this approach are still a question mark. One of the major limitations of current technologies, predicated on chromatography and mass spectrometry, is the limited measurable dynamic range i.e. typically 104 but in biological tissue and fluids the dynamic range can vary from 106 to 1010 [36]. This creates significant problems in terms of extent of coverage and limited sensitivity [36].

b. Also, the ability to integrate data from different platforms (Genomics, Proteomics, Transcriptomics, and Metabolomics) is very difficult because of the limited number of commercially available tools. Also there exists different types of customized approaching methods for biomarker analysis such as signal to noise ratio, t-tests, and Ecombo, (a whole genome comparative browser) [37] but there are no unifying standards.

Economics: Many authors have focused on the important -omics platforms used to undertake biomarker discovery. However, a number of authors have pointed out that “while there is genomics, transcriptomics, proteomics, metabolomics, pharmacogenomics and secretomics, the only really important -omics is ECON-omics”! [37,38]. The cost of developing these technologies for the biomarker discovery still remains very high and therefore it cannot yet be used as a routine diagnostic measure [39].

Errors in the assessment of a biomarker: One mistake often made when assessing the significance of biomarkers is to assume a strong association between a biomarker and disease by misinterpreting the statistical data and thus using the results inappropriately. Another potential misuse centers on the generalizability of biomarkers. There are at least five different “modifiers” that should be considered before using a biomarker for a specific patient: sex, co-morbidities, race, age and pathology [40]. An example of biomarker misuse would be to generalize results from a study of patients with cardiovascular disease to predict clinical outcomes in patients with no systemic disease.

Clinical use of biomarkers: Towards personalized medicine

The traditional treatment strategies used a “one size fits all” approach but as our knowledge of the underlying molecular causes of periodontitis continues to grow and along with the completion of human genome project, it is possible to develop personalized therapeutics that targets an individual patient’s needs based on the genotype. The concept of personalized medicine differs from that of evidence based medicine as the latter derives treatment decisions mainly from mean responses in studies designed either to minimize variability in response by inclusion and exclusion criteria or to tolerate the variability by increased size. But personalized medicine over turns this model to identify and exploit genetic differences among individuals within populations. In clinical practice, it means classifying individuals according to different biological pathways that may produce similar clinical signs and symptoms but produce different responses to disease initiators or treatments [41]. Accompanying the development of targeted therapeutics, there has been an increase in development of tests called companion diagnostics, or theranostics, that identify the disease or drug related biomarkers. Interest in their development has recently boosted, promoting the promise of personalized medicine. Examples of the companion diagnostics identifying disease related biomarkers include the Periodontal susceptibility test (PST®) which analyses two interleukins (IL-1α and IL-1β) genes for variations. Since IL-1 polymorphism is associated with increased of developing aggressive periodontitis [42] and peri-implantitis, this test can be used to identify high risk patients and modify treatment decisions accordingly. Another companion diagnostic kit available is the Periodontitis ++ (Autoimmu Diagnostika Aid, GMBH) which not only identifies five common bacterial pathogens (Actinobacillus actinomycetemcomitan, Bacteroides forsythus, Prevotella intermedia, Treponema denticola,Porphyromonas gingivalis) but also HLA DR4 antigen known to be commonly associated with aggressive periodontitis [43].

The AmpliChip CYP450, F. Hoffmann-La Roche Ltd, Switzerland test is the first FDA approved pharmacogenetic test to determine the genotype of the patient in terms of two cytochrome P450 enzymes: 2D6 and 2C19. They are responsible for the majority of the interindividual variability in the ability to metabolize drugs. If a drug, for example, phenytoin is given to the patient as a medication, and if the patient has reduced CYP2D6 or CYP2C19 activity, the patient will have elevated drug concentration in their body, and therefore severe side effects such as gingival overgrowth may occur. On the other hand, for the patient with increased activity, the drug concentration might be too low to have a therapeutic effect. So testing the phenotype of the patient is important to help determine the optimum dosage of the drug. The test analyses the DNA of a patient to determine the genotype, with the sample obtained from patients’ blood, buccal swab or saliva [44].

Conclusion

The use of biomarkers is growing, with new products being constantly brought in the market via diagnostics. Some of these biomarkers assist in identification, while others are channel either towards monitoring disease progression or evaluating the effectiveness of therapeutic options. These “novel biomarkers” have become the basis for preventive medicine, as it helps in promptly recognizing the disease or the risk of disease; and takes appropriate measures to prevent its progress. Furthermore, they offer another means for time-bound, rational drug design and development besides accelerating translational drug progress from animal to man. Biomarkers are now seen as the key to personalized medicine that helps in providing customized treatments to the individuals for highly efficient intervention in disease processes. However, in many cases, the evidence which supports the use of these new methods as opposed to traditional biochemical tests has not yet been demonstrated. Therefore, the ability to recognize, evaluate and understand the uses of existing and emerging biomarkers is an essential skill required of all biomedical health care professionals. Dentistry is at a crossroads at which we can either continue to use our traditional tools and traditional biomarkers to define oral disease or embrace what is emerging as the future tools of precision medicine.

References

  1. Baelum V, Lopez R. Periodontal epidemiology: towards social science or molecular biology? Community Dent Oral Epidemiol. 2004; 32: 239-249.
  2. The American Academy of Periodontology. The Pathogenesis of Periodontal Diseases; J Periodontol. 1999; 70: 457-470.
  3. Taba M Jr, Kinney J, Kim AS, Giannobile WV. Diagnostic biomarkers for oral and periodontal diseases. Dent Clin North Am. 2005; 49: 551-571.
  4. Haffajee AD, Cugini MA, Dibart S, Smith C, Kent RL Jr, Socransky SS. The effect of SRP on the clinical and microbiological parameters of periodontal diseases. J Clin Periodontol. 1997; 24: 324-334.
  5. Pear BB, Heath-Chiozzi M, Huff J. Clinical application of pharmacogenetics. Trends Mol Med. 2002; 7: 201-204.
  6. Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA. 1998; 279: 1200-1205.
  7. Garrod, Archibald E. The Incidence of Alkaptonuria: A Study in Chemical Individuality. Lancet. 1902; 2: 1616-1620.
  8. Sofaer JA. Genetic approaches in the study of periodontal diseases. J Clin Periodontol. 1990; 17: 401-408.
  9. Hodge PJ, Riggio MP, Kinane DF. Failure to detect an association with IL1 genotypes in European Caucasians with generalised early onset periodontitis. J Clin Periodontol. 2001; 28: 4306.
  10. Kumar M, Sarin S. Biomarkers of disease in medicine. Current Trends in Science. 2009: 403-417.
  11. Toomes C, James J, Wood AJ, Wu CL, McCormick D, Lench N, et al. Loss-of-function mutations in the cathepsin C gene result in periodontal disease and palmoplantar keratosis. Nat Genet. 1999; 23: 421-424.
  12. Noack B, Görgens H, Hempel U, Fanghänel J, Hoffmann T, Ziegler A, et al. Cathepsin C gene variants in aggressive periodontitis. J Dent Res. 2008; 87: 958-963.
  13. Blum SR. The Collagen Family. Cold Spring Harb Perspect Biol. 201; 1: 004978.
  14. López NJ, Jara L, Valenzuela CY. Association of interleukin-1 polymorphisms with periodontal disease. J Periodontol. 2005; 76: 234-243.
  15. Quappe L, Jara L, López NJ. Association of interleukin-1 polymorphisms with aggressive periodontitis. J Periodontol. 2004; 75: 1509-1515.`
  16. Ying L, Huang P, Lu X, Guan Dh, Man Y, Wei N, et al. The relationship between IL-1 gene polymorphism and marginal bone loss around dental implants. J Oral Maxillofac Surg. 2007; 65: 2340-2344.
  17. Machtei E, Oved-Peleg E, Peled M. Comparison of clinical, radiographic and immunological parameters of teeth and different dental implant platforms. Clin Oral Implants Res. 2006; 17: 658-665.
  18. Shao MY, Huang P, Cheng R, Hu T. Interleukin-6 polymorphisms modify the risk of periodontitis: a systematic review and meta-analysis. J Zhejiang Univ Sci B. 2009; 10: 920-927.
  19. Albuquerque CM, Cortinhas AJ, Morinha FJ, Leitão JC, Viegas CA, Bastos EM. Association of the IL-10 polymorphisms and periodontitis: a meta-analysis. Mol Biol Rep. 2012; 39: 9319-9329.
  20. Shimada Y, Tai H, Endo M, Kobayashi T, Akazawa K, Yamazaki K. Association of tumor necrosis factor receptor type 2 +587 gene polymorphism with severe chronic periodontitis. J Clin Periodontol. 2004; 31: 463-469.
  21. Zhang S, Barros SP, Moretti AJ, Yu N, Zhou J, Preisser JS, et al. Epigenetic Regulation of TNFA Expression in Periodontal Disease. J Periodontol. 2013; 84: 1606-1616.
  22. Ishida K, Kobayashi T, Ito S, Komatsu Y, Yokoyama T, Okada M, et al. Interleukin-6 gene promoter methylation in rheumatoid arthritis and chronic periodontitis. J Periodontol. 2012; 8: 917-925.
  23. Stefani FA, Viana MB, Dupim AC, Brito JA, Gomez RS, da Costa JE, et al. Expression, polymorphism and methylation pattern of interleukin-6 in periodontal tissues. Immunobiology. 2013; 218: 1012-1017.
  24. Oliveira NF, Damm GR, Andia DC, Salmon C, Nociti FH Jr, Line SR, et al. DNA methylation status of the IL8 gene promoter in oral cells of smokers and non-smokers with chronic periodontitis. J Clin Periodontol. 2009; 36: 719-725.
  25. Loo WT, Jin L, Cheung MN, Wang M, Chow LW. Epigenetic change in E-cadherin and COX-2 to predict chronic periodontitis. J Transl Med. 2010; 8: 110.
  26. Zhang S, Crivello A, Offenbacher S, Moretti A, Paquette DW, Barros SP. Interferon-gamma promoter hypomethylation and increased expression in chronic periodontitis. J Clin Periodontol. 2010; 37: 953-961.
  27. Zhang S, Barros SP, Niculescu MD, Moretti AJ, Preisser JS, Offenbacher S. Alteration of PTGS2 promoter methylation in chronic periodontitis. J Dent Res. 2010; 89: 133-137.
  28. Sim SC, Ingelman-Sundberg M. Pharmacogenomic biomarkers: New tools in current and future drug therapy. Trends Pharmacol Sci. 2011; 32: 72-81.
  29. Veronese ME, Mackenzie PI, Doecke CJ, McManus ME, Miners JO, Birkett DJ. Tolbutamide and phenytoin hydroxylations by cDNA-expressed human liver cytochrome P4502C9. Biochem Biophys Res Commun. 1991; 175: 1112-1118.
  30. Hassell TM. Stimulation and inhibition of fibroblast subpopulations by phenytoin and phenytoin metabolites, pathogenetic role in gingival enlargement. Pediatric Dental Journal. 1981; 3: 137-153.
  31. Pagliarini A, Stabellini G, Carinci F, Calura G, Tognon M, Evangelisti R. Heterogeneity of fibroblasts derived from human free and attached gingiva. Glycosaminoglycan synthesis and effects of phenytoin [PHT] treatment. J Oral Pathol Med. 1995; 24: 72-77.
  32. Kulasingam V, Diamandis EP. Strategies for discovering novel cancer biomarkers through utilization of emerging technologies. Nat Clin PractOncol. 2008; 5: 588-599.
  33. Patil PB, Patil BR. Saliva: A diagnostic biomarker of periodontal diseases. J Indian Soc Periodontol. 2011; 15: 310-317.
  34. Naylor S. Biomarkers: Current perspectives and future prospects. Expert Rev Mol Diasgn. 2003; 3: 525-529.
  35. Peng X, Wood CL, Blalock EM, Chen KC, Landfield PW, Stromberg AJ. Statically implications of pooling RNA samples for microarray experiments. BMC Bioinformatics. 2003; 4: 26.
  36. Jacobs JM, Adkins JN, Qian, WJ, Liu T, Shen Y, Camp DG, et al. Utilizing Human Blood plasma for proteomic biomarker discovery. J Proteome Res. 2005; 4: 1073-1085.
  37. Engels R, Yu T, Burge C, Mesirov JP, DeCaprio D, Galagan JE. Combo: a whole genome comparative browser. Bioinformatics. 2006; 22: 1782-1783.
  38. Buchanan J, Wordsworth S, Schuh A. Issues surrounding the health economic evaluation of genomic technologies. Pharmacogenomic. 2013; 14: 1833-1847.
  39. Ferber, G. Biomarkers and proof of concept. Methods Find Exp Clin Pharmacol. 2002; 24: 35-40.
  40. Glick M. The curious life of the biomarker. J Am Dent Asso. 2013; 144: 126-128.
  41. Kornman KS, Duff GW. Personalized medicine: will dentistry ride the wave or watch from the beach? J Dent Res. 2012; 91: 8-11.
  42. Kornman K, Crane A. The interleukin-1 genotype as a severity factor in adult periodontal disease. J Clin Periodontal. 1997; 24; 72-77.
  43. Firatli E, Kantarci A, Cebeci I, Tanyeri H, Sönmez G, Carin M, Tuncer O. Association between HLA antigens and early onset periodontitis. J Clin Periodontol. 1996; 23: 563-566.
  44. de Leon J, Susce MT, Johnson M, Hardin M, Maw L, Shao A, et al. DNA microarray technology in the clinical environment: The AmpliChip CYP450 test for CYP2D6 and CYP2C19 genotyping. CNS Spectr. 2009; 14: 19-34.

Download PDF

Citation:Luthra S, Grover HS and Maroo S. Genomic Biomarkers: Revolutionizing Diagnosis and Resolution of Periodontal Disease. J Dent & Oral Disord. 2016; 2(6): 1033. ISSN:2572-7710

Home
Journal Scope
Online First
Current Issue
Abstract Board
Instruction for Authors
Submit Your Article
Contact Us