Beukenlaan 137
5616 VD Eindhoven
The Netherlands
+31 85064 55 82
info@clinlabint.com
PanGlobal Media is not responsible for any error or omission that might occur in the electronic display of product or company data.
The human gene for anti-MĂźllerian hormone (AMH) was isolated and sequenced 20 years ago [1], with the first immunoassays developed in 1990 [2,3]. Since then, our understanding of this hormone has significantly increased, with most clinical use today focusing on womenâs reproductive health. AMHâs ability to reflect the number of small antral and pre-antral follicles present in the ovaries, and therefore the ovarian reserve, has led to AMH measurement being used in a wide array of clinical applications.
One of the first was as a tumour marker in the diagnosis and follow up of women with ovarian granulosa cell tumours (GCT) [4, 5]. More recently, with the dramatic improvements in the treatment of childhood cancers, attention is focused on AMH to assess the likelihood of gonadal damage and infertility after treatment. It is also being used to investigate the toxicity of different therapeutic regimens, in the choice of those treatments, and the prediction (and potential preservation) of fertility in young women and children following cancer therapy.
Sensitive diagnostic marker for GCT
GCT accounts for 2-3% of all ovarian tumours, with two distinct types: the juvenile and the adult form. The more common adult form generally presents in women at around 50 years. A majority have endocrine manifestations as a direct consequence of hormone secretion by the tumour [6].
GCTs have the potential to secrete estradiol, Inhibin (A and B) and AMH. Inhibin and AMH are the more useful biomarkers since estradiol is only produced in 50-60% of GCT patients and is dependent on stimulation by testosterone from adjacent theca cells. While serum total Inhibin is secreted in almost all GCT and has been shown to successfully detect recurrence following surgery, it is also increased in some epithelial ovarian tumours and fluctuates significantly within the menstrual cycle. AMH is more specific to GCT as expression is limited to ovarian granulosa cells and it does not change substantially over the menstrual cycle.
Although GCT is extremely rare, it is noted for its late recurrence, usually within four-six years, but can be up to 10-20 years after removal of the primary tumour. AMH disappears within days of removal of the ovaries [7] and, following tumour resection, a rise in AMH precedes clinical detection, making it an extremely sensitive marker for the early detection of tumour recurrence.
Laneâs 1999 study followed 56 patients post operatively and showed that AMH was useful in evaluating the completeness of tumour removal [4]. In addition, serial AMH measurements were able to detect recurrence on average three months prior to clinical detection. A second study, which followed 31 patients for up to seven years, confirmed these observations [5]. This group used an AMH assay 20 times more sensitive than previously used and, when comparing both assays found discrepant values in six out of 31 patients. The more sensitive assay accurately reflected the clinical situation and was elevated up to 16 months earlier in patients with tumour recurrence.
However, there is still insufficient published information on which to assess the sensitivity and specificity of AMH for the diagnosis of GCT. This is due to small patient numbers, the insensitivity of older assays and the lack of solid reference values in pre-menopausal women and children. The advent of more sensitive, fully automated assays will facilitate more robust studies.
Assessment of ovarian damage
The relationship between AMH and the number of small growing follicles (and therefore the number of primordial follicles or ovarian reserve) makes it useful for assessing the gonadal toxicity of cancer therapy and loss of ovarian reserve. Levels fall rapidly with the onset of cancer treatment, with subsequent recovery dependant on degree of ovarian damage. AMH appears to identify which treatments may spare the ovaries, or are most toxic to them, and may give clinicians additional information to direct therapeutic choices in children and women of childbearing age with cancer.
Radiotherapy is a well-known cause of ovarian damage, even at low radiation levels. Women who have undergone pelvic or total body irradiation are likely to have low or undetectable AMH levels [9, 10]. The gonadal toxicity of alkylating agents is also well established. In a study involving young women with lymphoma, those receiving alkylating agents showed little or no recovery in AMH levels following treatment whereas those receiving alternative chemotherapy showed good recovery.
Childhood cancer and fertility
Childhood cancer treatment has improved dramatically with survival rates of more than 90%. However, the consequences of treatment may be permanent damage to the ovaries, affecting fertility. AMH is detectable in females of all ages rising steadily throughout childhood. Several studies have confirmed its role as a clinically useful marker to assess impairment of ovarian reserve in those receiving treatment for cancer [11, 12, 13].
Brougham showed that AMH decreased during chemotherapy in both prepubertal and pubertal girls, becoming undetectable in 50% of patients; recovery occurred in the low to medium risk groups after completion of treatment, yet remained undetectable in the high risk group. Inhibin B was undetectable in most patients before treatment and FSH showed no relationship with treatment. Thus AMH indicates a more useful assessment of residual ovarian reserve, revealing partial loss or ovarian failure.
It is clear that a woman can suffer a significant loss of ovarian reserve without any lasting effects on her fertility, for example following removal of an ovary. For survivors of childhood cancer this may mean that only a substantial loss of ovarian reserve would have a clinical impact. Indeed, recent work has shown that there is a high number of successful pregnancies in lymphoma survivors, despite low AMH levels [14]. In a study of 84 childhood cancer survivors they achieved pregnancy rates similar to controls despite impaired ovarian reserve [15]. However, a 10-year follow up study of childhood cancer survivors, now in their 30s, showed that the percentage of childless women in this group was greater than in the normal Danish population, particularly in the group of women who received the most gonadotoxic treatment burden. Their pregnancy rate and outcome was especially poor [16]. The truth is difficult to discern on current evidence and more work is required on long term follow up, with fertility and age at menopause as end points.
The real value of measuring AMH in young women surviving cancer would be to forecast long-term reproductive outcome and take steps to preserve their fertility.
Reproductive outcomes in adult women
The same fertility concerns exist for women of childbearing age. Using AMH values to assess ovarian reserve and individualize risk, more invasive methods of fertility preservation may be appropriate for women with a low AMH, while those with high values for their age may decide to start cancer treatment without delay.
Most evidence comes from breast cancer studies and is based on the assumption that a woman with a higher pre-treatment AMH before chemotherapy will be more likely to retain ovarian function. A prospective study in women with newly diagnosed breast cancer linked high levels of AMH detected before treatment with retaining long-term ovarian function five years after surgery [17]. Pretreatment serum AMH was seen to be markedly higher in women who continued to have menses. The predictive value of AMH for post-chemotherapy ovarian function has subsequently been confirmed [18] allowing the development of prediction tools combining age and AMH [18].
Individualizing breast cancer adjuvant chemotherapy
Adjuvant endocrine therapy has been shown to reduce the likelihood of reocurrence and improve overall survival rates in hormone receptor-positive (HR-positive) breast cancer. However, it appears that ovarian function after chemotherapy has direct implications on the choice of therapy. Aromatase inhibitors (AIs) are more effective in postmenopausal women than tamoxifen [19]. However, in premenopausal women, AIs may cause a rise in estrogen levels due to reactivation of ovarian function. Consequently, even in women who have developed chemotherapy-induced ovarian failure, tamoxifen is the standard of care [20, 21].
It has been suggested that all women who are premenopausal prior to chemotherapy, even those in their late 40s and early 50s, should be treated with adjuvant tamoxifen therapy or, if they are going to receive an aromatase inhibitor, should have their ovaries removed or chemically suppressed [22]. For the latter group, these strategies are invasive and are associated with increased side effects. Consequently, being able to predict permanent ovarian failure using information other than the patientâs age is relevant.
Data from recent studies [8, 17] suggest that pre-chemotherapy assessment of serum AMH concentrations, possibly in combination with inhibin B, may provide important information about the likelihood of developing permanent ovarian failure with chemotherapy. In addition, this could help identify a patient population in which it would be safe to treat with upfront AI monotherapy. The expanding number of studies available all add to our understanding of the role of AMH in ovarian function, its ability to predict a womanâs ovarian reserve for her fertility and the impact of cancer treatment on reproductive health.
References
1. Cate RL, Mattaliano RJ, Hession C, et al. Isolation of the bovine and human genes for MĂźllerian inhibiting substance and expression of the human gene in animal cells. Cell 1986; 45, 685-698.
2. Hudson PL, Dougas I, Donahoe PK, et al. An immunoassay to detect human MĂźllerian inhibiting substance in males and females during normal development. J Clin Endocrinol Metab. 1990; 70, 16-22.
3. Josso et al. An enzyme linked immunoassay for anti-mĂźllerian hormone: a new tool for the evaluation of testicular function in infants and children. JCEM 1990; 70, 23-27.
4. Lane AH, Lee MM, Fuller AF Jr, et al. Diagnostic utility of MĂźllerian inhibiting substance determination in patients with primary and recurrent granulosa cell tumors. Gynecol Oncol 1999; 73, :51â55.
5. Long WQ, Ranchin V, Pautier P, et al. Detection of minimal levels of serum anti-MĂźllerian hormone during follow-up of patients with ovarian granulosa cell tumor by means of a highly sensitive enzyme-linked immunosorbent assay. J Clin Endocrinol Metab 2000; 85, 540â544.
6. Bjorkholm E, Silfversward C. Prognostic factors in granulosa-cell tumors. Gynecol Oncol. 1981;11, 261â274.
7. LaMarca A, De Leo V, Giulini S, et al. Anti-MĂźllerian hormone in premenopausal women and after spontaneous or surgically induced menopause. J Soc Gynecol Invest 2005; 12, 545â548.
8. Henry, NL, Xia R, Schott AF, McConnell D, et al. Prediction of Postchemotherapy Ovarian Function Using Markers of Ovarian Reserve. The Oncologist 2014; 19, 68â74.
9. Lie Fong S, Laven JS, Hakvoort-Cammel FG, et al. Assessment of ovarian reserve in adult childhood cancer survivors using anti-Mullerian hormone. Hum Reprod 2009;24, 982â990
10. Gracia CR, Sammel MD, Freeman E, et al. Impact of cancer therapies on ovarian reserve. Fertil Steril 2012; 97, 134â140 e131.
11. Bath LE, Wallace WH, Shaw MP, et al. Depletion of ovarian reserve in young women after treatment for cancer in childhood: detection by anti-MĂźllerian hormone, inhibin B and ovarian ultrasound. Hum Reprod 2003; 18, 2368â2374.
12. van Beek RD, van den Heuvel-Eibrink MM, Laven JS, et al. Anti-MĂźllerian hormone is a sensitive serum marker for gonadal function in women treated for Hodgkinâs lymphoma during childhood. J Clin Endocrinol Metab 2007; 92, 3869â3874.
13. Brougham MF, Crofton PM, Johnson EJ, et al. Anti-MĂźllerian hormone is a marker of gonadotoxicity in pre- and postpubertal girls treated for cancer: a prospective study. J Clin Endocrinol Metab 2012; 97, 2059â2067.
14. Janse F, Donnez J, Anckaert E, et al. Limited value of ovarian function markers following orthotopic transplantation of ovarian tissue after gonadotoxic treatment. J Clin Endocrinol Metab 2011; 96, 1136â1144.
15. Dillon KE, Sammel MD, Ginsberg JP, et al. Pregnancy After Cancer: Results From a Prospective Cohort Study of Cancer Survivors. Pediatr Blood Cancer. 2013 Dec; 60(12), 2001-6.
16. Nielsen SN, Andersen AN, Schmidt KT, et al. A 10-year follow up of reproductive function in women treated for childhood cancer. Reprod Biomed Online 2013; 27, 192â200.
17. Anderson RA, Cameron DA. Pretreatment serum anti-mĂźllerian hormone predicts long-term ovarian function and bone mass after chemotherapy for early breast cancer. J Clin Endocrinol Metab 2011; 96, 1336â1343.
18. Anderson RA, Rosendahl M, Kelsey TW, et al. Pretreatment anti-MĂźllerian hormone predicts for loss of ovarian function after chemotherapy for early breast cancer. Eur J Cancer 2013;49, 3404â3411.
19. Burstein HJ, Prestrud AA, Seidenfeld J et al. American Society of Clinical Oncology clinical practice guideline: Update on adjuvant endocrine therapy for women with hormone receptor-positive breast cancer. J Clin Oncol 2010; 28, 3784â3796.
20. Smith IE, Dowsett M, Yap Y-S et al. Adjuvant aromatase inhibitors for early breast cancer after chemotherapy-induced amenorrhoea: Caution and suggested guidelines. J Clin Oncol 2006; 24, 2444â2447.
21. Burstein HJ, Mayer E, Patridge AH et al. Inadvertent use of aromatase inhibitors in patients with breast cancer with residual ovarian function: Cases and lessons. Clin Breast Cancer 2006;7, 158â161.
22. Henry NL, Xia R, Banerjee M et al. Predictors of recovery of ovarian function during aromatase inhibitor therapy. Ann Oncol 2013; 24, 2011â2016.
The author
Sherry Faye, PhD
Director, Global Scientific Affairs,
Beckman Coulter Diagnostics
Brea, CA, USA
Pharmacogenomic research has been active in the area of hepatitis C virus infection. Both viral and host polymorphisms are discussed in this review to describe how the genotype information helps predict response to conventional peginterferon alfa and ribavirin therapy as well as more recently recommended direct-acting antivirals, simeprevir and sofosbuvir.
by M. Kawaguchi-Suzuki and Dr R. F. Frye
Hepatitis C virus infection
Hepatitis C virus (HCV) chronically infects 170 million people worldwide [1]. After exposure to HCV, some patients may experience fever, fatigue, dark urine, clay-coloured stool, abdominal pain, loss of appetite, nausea, vomiting, joint pain or jaundice [2]. However, the majority of patients are asymptomatic and do not seek medical attention, leading to the chronic infection rate of 75â85% [2]. HCV infection is a significant burden to society, not only because of the prevalence but also because the infection can lead to severe complications and mortality. Patients infected with HCV can develop chronic liver disease, progressing to advanced fibrosis and cirrhosis and eventually to hepatocellular carcinoma [1]. Additionally, HCV infection is the primary indication for liver transplantation in developed countries [1].
HCV is a blood-borne 9.6 kb positive-sense, single-stranded RNA virus [1, 2]. Typically, the first screening method used to diagnose HCV is the detection of anti-HCV antibodies [3]. Definitive diagnosis of HCV infection is then made by measurements of HCV RNA or âviral loadâ, with a sensitive molecular method having a lower limit of detection <15 IU/mL [3, 4]. Once a decision to treat has been made, the therapeutic goal is to achieve virologic cure or sustained virologic response (SVR) defined as undetectable HCV RNA 12 weeks after the completion of therapy [4]. Recent advancements in HCV treatment now make HCV curable for more patients, which will reduce mortality and liver-related health adverse consequences.
Therapy for HCV infection
Since the identification of HCV in 1989, various treatment regimens have been used to treat HCV infection [1]. Interferon therapy was the first treatment against HCV and then combination therapy with ribavirin (RBV) became available in 1998 [1]. After the introduction of the pegylated formulation of interferon or peginterferon alfa (PEG) in 2001, the dual therapy of PEG and RBV has been the standard care for a decade, until the recent approval of direct-acting antivirals (DAAs) [1]. The first-generation DAAs are the protease inhibitors boceprevir and telaprevir. Subsequently, another protease inhibitor, simeprevir, and a polymerase inhibitor, sofosbuvir, were introduced. The three protease inhibitors were approved in combination with PEG and RBV. However, PEG-free regimens became an option for select patients with the approval of sofosbuvir.
Although the likelihood of achieving SVR has improved with each advance in treatment, HCV infection is a therapeutic area in which large inter-patient variability in response has been observed. In order to predict treatment response and to tailor therapy for individual patients infected with HCV, the extent to which pharmacogenomic biomarkers explain this variability has been examined.
Pharmacogenomics: viral polymorphism
HCV is categorized into genotypes 1â7 and further classified into subtypes a, b, etc., based on sequence divergence [1]. The observed HCV genotype depends largely on the geographic location; genotype 1 accounts for 70% of cases in the Americas, 50â70% of cases in Europe, and 75% of cases in Japan, with genotypes 2 and 3 being the next most prevalent [1]. In contrast, genotypes 3 and 6 have been widely identified in South and Southeast Asia, while genotypes 4 and 5 are most commonly observed in Africa [1]. Genotype 7 was most recently discovered, but its clinical importance has not yet been determined [1]. Identification of the HCV genotype is important for HCV-infected patients because treatment choice, therapy duration, and treatment response depend on the viral genotype. The first-generation DAAs and simeprevir are only indicated for genotype 1 infection. However, sofosbuvir has pan-genotypic activity and is approved for the treatment of genotypes 1, 2, 3, and 4 [5].
Before the approval of the use of DAAs, HCV therapy targeted the host immune system with the use of PEG. However, since therapy now directly targets the virus itself, various viral polymorphisms that may confer treatment resistance have been reported. The most notable one is the Q80K polymorphism observed in HCV genotype 1a [6]. Findings from the phase 3 QUEST-1, QUEST-2, and PROMISE trials are shown in Figure 1 [6]. QUEST-1 and QUEST-2 were conducted among treatment-naĂŻve patients, whereas PROMISE was a study in patients who relapsed after previous treatment with an interferon-based regimen.
In general, HCV genotype 1a was considered less susceptible to the treatment, but when the data were analysed based on Q80K polymorphism, the SVR rates in genotype 1a were similar to those in genotype 1b if the Q80K polymorphism was absent. However, the SVR rates turned out to be even lower when the polymorphism was present. Based on these data, both prescribing information and clinical guidelines suggest alternative therapy if the Q80K polymorphism is detected in HCV genotype 1a infection [6]. Consequently, Q80K polymorphism testing is recommended by the clinical guidelines in all patients before the initiation of simeprevir, PEG, and RBV triple regimen [4].
Pharmacogenomics: host polymorphism
Earlier, various genome-wide association studies and candidate gene studies found two single nucleotide polymorphisms (SNPs) in the host were associated with SVR in HCV genotype 1 infection after PEG and RBV therapy [7]. The two SNPs, rs12979860 and rs8099917, are located in IFNL3 gene (previously called IL28B) [7]. Rs12979860 CC and rs8099917 TT genotypes have been considered as favourable response genotypes, with SVR rates of around 80% achieved in genotype 1 infection, whereas rs12979860 minor T allele or rs8099917 minor G allele carriers had lower SVR rates of about 20% [8]. SVR rates tend to be higher in HCV genotype 2 and 3 infections with PEG and RBV therapy, compared to those in genotype 1 therapy, and the association of these two SNPs with SVR has not been as strong in genotype 2 and 3 infections [7]. However, similar association of the two IFNL3 SNPs with SVR to that in genotype 1 infection has been shown in genotype 4 infection [7]. Although data has been scarce in other rare genotype infections, the IFNL3 genotype has been one of the strongest predictors of SVR with PEG and RBV therapy [7, 8]. The exact mechanism by which the IFNL3 SNPs affect the phenotype has not been fully elucidated, but baseline differences in the expression level of interferon-stimulated genes have been proposed [9]. Data has been collected with both IFNL3 SNPs, but rs12979860 is more commonly used if a single SNP has to be chosen for research or clinical purpose.
The association of treatment response with the IFNL3 genotype has also been observed with interferon-based DAA therapies. Currently, treatment with boceprevir or telaprevir is not recommended by the guidelines, and most commonly used regiments include simeprevir and/or sofosbuvir [4]. Figure 2 describes SVR rates in treatment-naĂŻve and treatment-experienced patients who were treated with simeprevir or sofosbuvir combined with PEG and RBV as indicated in the prescribing information [5, 6]. Higher SVR rates were consistently observed in patients with the rs12979860 CC genotype, compared to the T allele carriers. However, it should be noted that the difference in SVR rates between the IFNL3 genotypes was comparatively modest with the addition of a DAA to the conventional PEG and RBV therapy. In addition, no significant difference was observed in SVR rates based on the IFNL3 genotype in patients infected with HCV genotype 2 or 3 after an interferon-free regimen of sofosbuvir and RBV [10].
Summary and future directions
Large inter-patient variability exists in response to PEG and RBV therapy, and the IFNL3 genotype has been demonstrated as one of the strongest predictors for SVR, especially in HCV genotype 1 infection. This trend has also been observed in interferon-based DAA therapies. However, if an interferon-free regimen becomes more readily available in the future, with the potential for SVR rates to approach 100%, the IFNL3 genotype may no longer hold clinical utility. However, IFNL3 genotype may still need to be tested during drug development to ensure that investigational agents will have efficacy in patients carrying the variant allele previously associated with an unfavourable treatment response. Additionally, when PEG is eliminated from treatment regimens and only DAAs are combined, cross-resistance may become a concern in the future, especially in patients who failed a DAA regimen previously. Various viral polymorphisms have been detected in protease inhibitors, and cross-resistance can be an issue in this drug class [6, 11, 12]. Viral polymorphisms may play a bigger role and need to be monitored for the future.
References
1. Scheel TK, Rice CM. Understanding the hepatitis C virus life cycle paves the way for highly effective therapies. Nat Med. 2013; 19(7): 837â849.
2. Centers for Disease Control and Prevention. Hepatitis C information for health professionals. 2013; http://www.cdc.gov/hepatitis/hcv/. Accessed January 11, 2014.
3. EASL. EASL Clinical Practice Guidelines: Management of hepatitis C virus infection. J Hepatol. 2014; 60(2): 392â420.
4. AASLD, IDSA, IASâUSA. Recommendations for testing, managing, and treating hepatitis C. 2014; http://www.hcvguidelines.org/.
5. Sovaldi (sofosbuvir) package insert. Gilead Sciences, Inc. 2013.
6. Olysio (simeprevir) package insert. Janssen Therapeutics. 2013.
7. Kawaguchi-Suzuki M, Frye RF. The role of pharmacogenetics in the treatment of chronic hepatitis C infection. Pharmacotherapy. 2014; 34(2): 185â201.
8. Pacanowski M, et al. New genetic discoveries and treatment for hepatitis C. JAMA. 2012; 307(18): 1921â1922.
9. Cariani E, et al. Translating pharmacogenetics into clinical practice: interleukin (IL)28B and inosine triphosphatase (ITPA) polymophisms in hepatitis C virus (HCV) infection. Clin Chem Lab Med. 2011; 49(8): 1247â1256.
10. Jacobson IM, et al. Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. N Engl J Med. 2013; 368(20): 1867â1877.
11. Victrelis (boceprevir) package insert. Merck & Co., Inc. 2013.
12. Incivek (telaprevir) package insert. Vertex Pharmaceuticals 2013.
The authors
Marina Kawaguchi-Suzuki PharmD, BCPS; Reginald F. Frye* PharmD, PhD, FCCP
Department of Pharmacotherapy and Translational Research,
University of Florida College of Pharmacy, Gainesville,
FL 32610-0486, USA
*Corresponding author
E-mail: frye@cop.ufl.edu
Inherited skin diseases can be difficult to assess clinically and often diagnosis relies on multiple laboratory investigations. Traditionally, examination of skin biopsies is followed by biochemical testing and Sanger sequencing of genomic DNA. This approach is labour-intensive, costly and time-consuming. The advent of next-generation sequencing (NGS) methods provides an alternative or complementary approach to making highly accurate diagnoses, but is not without its own challenges.
by J. Lee, Dr A. Salam, Dr T. Takeichi and Prof. J. A. McGrath
Background
The identification of pathogenic mutations in monogenic diseases represents one of the major challenges, and fundamental goals, of early 21st Century human genetics. Most genetic diseases are rare, clinically heterogeneous, and difficult to diagnose â a task made more challenging by disparity in genotypeâphenotype correlations, inter- and intra-familial variability, and well as mosaic patterns of disease. It is these hurdles that have led to the advent of Next-Generation DNA Sequencing (NGS); a group of technologies that can improve the speed, accuracy, and cost-efficiency of genetic sequencing, while simultaneously mapping normal variation, and thus furthering our understanding of human genetics in both health and disease. Inherited skin diseases encompass a collection of over 500 clinical entities â with variable structural or inflammatory manifestations that can also affect hair, nails, teeth and certain mucosal surfaces [1]. Individually these disorders are uncommon, but collectively they generate a significant health burden and many diagnostic conundrums.
Traditional approaches to the diagnosis of inherited skin diseases
For patients with inherited skin disorders, the traditional approach to diagnosis is to document a comprehensive patient history, including recording accurate family pedigrees, and noting any consanguinity. The clinician will then go on to perform a physical examination, take clinical photographs, and order laboratory investigations, which often include a skin biopsy. Light microscopy is usually uninformative, and the skin may need to be examined by transmission electron microscopy and immunohistochemistry. Additional blood or urine samples may be need for further diagnostic biochemical studies. Changes in skin structure or protein expression may provide clues to candidate genes, for which polymerase chain reaction primers can be designed and used for Sanger sequencing of genomic DNA. This âcandidate geneâ approach has proved very useful for several autosomal recessive inherited skin diseases, but is typically unhelpful in most dominant diseases or in those with more subtle changes in skin morphology. Cue the advent of NGS technologies and a different approach to diagnostics, where the challenge in genetic discovery shifts away from the generation of data, to the filtering of relevant data [2, 3].
The impact of NGS
NGS encompasses a number of new technologies that vary in their sequencing protocols, thus determining the type of data produced. The approaches taken vary in template preparation, sequencing and imaging, genome alignment and assembly methods. The methodology is therefore also known as high throughput or massively parallel sequencing due to the ability of NGS to process large volumes of genetic data in a short time, in stark contrast to individual gene screening with Sanger sequencing. Whole-genome sequencing (WGS) and whole-exome sequencing (WES) are the two most commonly used NGS techniques. WGS has the ability to sequence an individualâs entire genome, but at the expense of speed and cost. In contrast, WES uses an array to capture protein-coding regions of the human genome, encompassing ~21,000 genes, which make up less than 2% of the genome. Compared with the 2â3 million variants generated by WGS, the data from WES typically reveals around 25,000 variants. Nevertheless, WES is a more economical option than WGS because ~85% of the pathogenic mutations in monogenic diseases are predicted to be in exons. The plethora of data then has to be filtered, with any potentially disease variant with evidence for causality established (Fig. 1). This process often involves the filtering of variants through databases of previously identified sequences, and cross referencing with known biological or genetic databases, for which considerable bioinformatics support is required: a single WES run can generate one terabyte of data.
Whole-exome sequencing: the possible advantages
The challenge
The key questions for WES in the diagnosis of inherited skin diseases are as follows. (1) Are the new technologies better than what already exist for diagnosing known diseases? (2) Can the new technologies be helpful in resolving unknown diagnoses or discovering new clinical entities? (3) Can the new technologies be introduced into clinical work and overcome any practical obstacles? Emerging data indicate a resounding yes to the first two questions, although the third remains work in progress [4].
Breadth of cover
WES encompasses most of the coding regions of the genome, whereas Sanger sequencing targets a predetermined gene, or part of a gene, between specially designed primers. WES is also efficient for sequencing large genes, such as COL7A1, which encodes type VII collagen. This gene, which is mutated in the blistering disease, dystrophic epidermolysis bullosa, is composed of 118 exons. Conventional Sanger sequencing approaches are based on designing ~72 primer pairs to amplify the COL7A1 exons and flanking introns. Thus the Sanger sequencing approach is therefore laborious and expensive, particularly as COL7A1 contains few recurrent mutations and the gene needs to be screened in its entirety to identify pathogenic mutations.
Genetic diagnosis
WES has emerged as an invaluable tool where a patientâs clinical diagnosis is unclear or erroneous. In this situation, Sanger sequencing of multiple candidate genes is destined to failure and to exhaust both time and resources. WES, on the other hand, can identify known variants in order to make a genetic diagnosis that was not initially considered, as has been demonstrated for subtypes of epidermolysis bullosa, and other inherited skin diseases [5, 6]. Indeed, WES has been used to accurately diagnose inherited skin diseases without any a priori clinical information [7]. The rationale is that more accurate and timely diagnoses offered by WES will allow for earlier targeted therapy and ultimately improved patient care.
Genetic discovery
The value of WES in genetic discovery is evident in the number of inherited skin diseases whose original genetic basis has been informed by WES. Recent examples include the discovery of inherited skin and bowel inflammation resulting from mutations in ADAM17 and EGFR [8, 9]. Given the protean nature of inherited skin diseases, many mutations cannot be anticipated based on clinical phenotype and initial investigations, leaving no candidate gene targets for Sanger sequencing. One pertinent example of the completely unexpected candidate gene is the identification of mutations in EXPH5 [10], which encodes a GTPase effector protein, exophilin-5, in a form of intra-epidermal epidermolysis bullosa â a disease that usually arises as a genetic disorder of keratin. WES is therefore superior to Sanger sequencing in the diagnosis of both novel and genetically heterogeneous conditions.
Cost efficiency
The cost of DNA sequencing has reduced by around 100,000-fold over the last 20 years. Although the technique remains relatively expensive at present (~ÂŁ900 per sample at Kingâs College London, 2014 prices), further cost reductions are expected that will soon make WES a more economically viable option than Sanger sequencing, for all but a few disorders in which there are recurrent mutations in a small number of small genes. Even at current costs, however, WES already has advantages over Sanger sequencing for some genes, such as COL7A1, for which the cost of Sanger sequencing is ~ÂŁ1000 (or greater) in the small number of laboratories that undertake sequencing of this gene.
Considering the patient
The diagnosis of many inherited skin disorders often relies on invasive investigations such as sampling a small piece of skin (punch or ellipse biopsy) (Fig. 2). The procedure involves injection of local anesthetic, which can be painful, and the wound usually heals with a small but evident scar. Occasionally, skin biopsy sites can be complicated by bleeding or infection. WES can be performed using DNA extracted from blood, saliva or tissue samples, and although Sanger sequencing can also be performed on similar templates, for many patients, a skin biopsy would have been necessary to determine the gene(s) for sequencing. Thus WES typically offers a less-invasive approach for the patient.
Variant mapping
Aside from discovering genes and pinpointing mutations in inherited skin diseases, WES also generates a huge amount of other data that can be used to map genetic variation. In the longer term, the dissection of bioinformatics data will lead to a better understanding of the implications of certain variants, refining genotypeâphenotype correlation, thus providing insight into individual prognosis, and allowing stratified or personalized medicine and therapeutics.
Whole-exome sequencing: the possible disadvantages
Data analysis
The large quantity of sequencing data generated by WES is potentially also a disadvantage. Before WES can be used in routine clinical practice, fast and efficient filtering techniques must exist to allow clinicians and non-geneticists to interpret WES data and to extract the relevant information in order to manage their patientâs needs. But the plethora of data generated by WES also provides considerably more information beyond the pathogenic mutation itself, including several co-incidental potentially damaging mutations (known as âincidental findingsâ) that are completely unconnected to the primary disease being investigated. What should diagnosticians do with this information? Does it make a difference if the implications are clinically actionable or not? There are clearly several unresolved issues.
Accuracy of data
Given the volume of data produced by WES, it is inevitable that some false positive variants are identified. Most laboratories therefore still elect to confirm mutations via an alternative sequencing platform, generally Sanger sequencing, which is therefore a significant barrier to the routine use of WES in diagnostics. From a technical perspective, NGS methods still need to be improved to cover important regulatory elements such as promoters and enhancers, and poorly annotated parts of the genome. Moreover, if WES is to become a routine diagnostic technique, standardized operating procedures and protocols must be created and implemented. For inherited skin disease diagnostics there would also need to be a realignment of technical wet lab skills (skin microscopy) in favour of computer database and in silico work.
Time to diagnosis
Perhaps the biggest challenge for WES, however, lies in the time it takes to process and analyse a case. For many inherited skin diseases, a rapid diagnosis is often very important to optimize clinical management, for example in neonates with suspected epidermolysis bullosa. The diagnostic approach using skin biopsy assessment followed by Sanger sequencing of candidate genes (implicated by skin biopsy) allows for possible diagnoses to be made within 2 to 3 days. In contrast, the quickest time that WES could be completed (at present) would be a minimum of 5 days, although in practice WES often takes considerably longer to complete and analyse. New platforms to shorten WES protocols are in development, but only when more rapid sample analysis is feasible in a diagnostic lab setting can one really begin to think about wholesale change of diagnostic practice.
Conclusion
Since 2011, WES has proven to be a valuable asset in the diagnosis and discovery of inherited skin diseases. But the adoption of WES into clinical diagnostics diagnosis is still being refined and piloted. WES techniques are constantly being improved to become more accurate, quicker and cost-effective, while enrichment methodologies and sequencing technology become more reproducible and standardized. This progress may allow WES to function independently as the stand alone diagnostic and discovery tool in genetics, negating the need for Sanger sequencing to confirm WES findings. However, as our understanding of the role of non-coding DNA in molecular biology grows, and as WGS is further refined, WES is at risk of being superseded by newer NGS techniques both for genetic discovery diagnostics and prognostics. Innovation looms, but ever it was in molecular genetics.
References
1. Leech SN, Moss C. Br J Dermatol. 2007; 156: 1115â1148.
2. Metzker ML. Genome Res. 2005; 15: 1767â1776.
3. Metzker ML. Nat Rev Genet. 2010; 11: 31â46.
4. Cho RJ, et al. J Invest Dermatol. 2012; 132(E1): E27â28.
5. Takeichi T, et al. Br J Dermatol. 2014; doi: 10.1111/bjd.13190. [Epub ahead of print]
6. Salam A, et al. Matrix Biol. 2013; 33: 35â40.
7. Takeichi T, et al. Exp Dermatol. 2013; 22: 825â831.
8. Blaydon DC, et al. N Engl J Med. 2011; 365: 1502â1508.
9. Campbell P, et al. J Invest Dermatol. 2014; doi: 10.1038/jid.2014.164. [Epub ahead of print].
10. McGrath JA, et al. Am J Hum Genet. 2012; 91: 1115â1121.
The authors
John Lee, Amr Salam BSc, MBChB, MRCP(UK), Takuya Takeichi MD PhD, John A McGrath* MD FRCP
St Johnâs Institute of Dermatology, Kingâs College London (Guyâs Campus), London, UK.
*Corresponding author
E-mail: john.mccgrath@kcl.ac.uk
Although formally defined as recently as the early 2000s, biomarkers have quickly begun to gain acceptance in clinical practice. Many experts believe they will become an indispensable tool for the diagnosis and management of a wide variety of medical conditions in the near future.
Cardiovascular disease now a global priority
One of the priority applications for biomarkers is likely to be for cardiovascular diseases (CVD) – the leading cause of mortality and disability in the Western world. In Europe, CVD causes 1.9 million deaths a year, while the toll in the US is about 1 million.
The prevalence of CVD is also increasing rapidly in newly industrializing countries, especially among the more affluent urban populations adopting Western lifestyles. Indeed, âCVD is now more numerous in India and China than in all economically developed countries in the world added together.â
Mapping the disease progression pathway
It has, for some time, been accepted that CVD follows a relatively clear-cut pathway from subclinical to overt status. The Multi-Ethnic Study of Atherosclerosis (MESA), sponsored in the year 2000 by the US National Institutes of Health, has been seeking to assess the characteristics of subclinical CVD and means to predict its progression to clinically overt cardiovascular disease. More recently, in 2010, Spainâs Banco Santander and the Istituto de Salud Carlos III launched a similar effort in Europe called PESA (Progression of Early Subclinical Atherosclerosis).
Such efforts are targeted at providing clinicians with tools to help assess CVD and identify vulnerable, at-risk patients. In many respects, they complement the worldâs most ambitious effort in the area, the Framingham Heart Study, which began in 1948 in a town in Massachusetts in the US with 5,209 adult subjects. The Study, which has now enrolled its third generation of participants, has resulted in the publication of over 1,000 medical papers. It has also provided many commonplace tools for the contemporary understanding of CVD, including the impact of smoking, diet and exercise, medications such as aspirin etc. – as well as the term ârisk factorâ.
The Framingham project: clarifying the role of biomarkers
Biomarkers began to be part of the Framingham project in the 2000s, although initial results were unclear. For instance, enthusiasm about elevated levels of the inflammation marker C-reactive protein (CRP) as an independent risk factor for future CVD events were dispelled in a 2005 study supported by the Framingham sponsors.
In September 2012, a study in the American Heart Associationâs journal âCirculationâ pointed to one reason for such conflicting assessments, namely the âlack of cardiovascular specificityâ in many of the new biomarkers. The authors sought to address such limitations by studying three key CVD biomarkers (soluble ST2, growth differentiation factor-15 and high-sensitivity troponin I) in almost 3,500 patients. The findings were conclusive: âMultiple biomarkers of cardiovascular stress,â they said âadd prognostic value to standard risk factors for predicting death, overall cardiovascular events, and heart failure.â
In 2014, another study of 2,680 Framingham participants sought to associate circulating biomarkers with The American Heart Association Cardiovascular Health score (CVH score). The authors concluded there was an âinverse associationâ between ideal CVH and CVD incidence, and that this was partly attributable to its âfavourable impact on CVD biomarker levels and subclinical disease.â The list of CVD biomarkers in the 2014 study includes natriuretic peptides (N-terminal pro-atrial and B-type natriuretic peptide), plasminogen activator inhibitor-1, aldosterone, C-reactive protein, D-dimer, fibrinogen, homocysteine and growth differentiation factor-15.
Identification of at-risk patients
One of the most promising biomarkers seems to be cardiac troponin, first identified in the early 1990s. Changes in cardiac troponin T (cTnT) levels over time appear to correlate with heart failure risk, especially in a major study of elderly subjects.
The potential of circulating cTnT may also extend beyond the heart failure setting. Some argue that circulating cTnT is representative of subclinical myocardial dysfunction. In the general population, studies show that elevated cTnT is associated with subclinical cardiac injury, and marks an increased risk for structural heart disease and all-cause mortality.
Other studies have found that myeloperoxidase (MPO) and high-sensitivity C-reactive protein (hsCRP) in apparently healthy populations can predict risk of coronary disease, allowing for early preventative treatment. Together, MPO and C-reactive protein have also shown promise in prognostic risk assessment for patients with systolic heart failure.
Enabling targeted and timely treatment
While screening the general population is bound to draw considerable attention, the more immediate application of CVD biomarkers is to enable treatment in a risk-stratified and timely fashion.
One of the biggest challenges faced by physicians is to differentiate between patients with unstable angina and acute myocardial infarction (AMI) in an emergency setting. Here too cTnT – as well as cardiac troponin I (cTnI) – have catalysed some of the greatest excitement, due to their high sensitivity and specificity for cardiac damage.
In 2007, the US National Academy of Clinical Biochemistry Laboratory Medicine Practice recommended the use of cardiac troponin as a âpreferredâ biomarker for MI diagnosis, in conjunction with clinical evidence of myocardial ischemia. Creatine kinase-MB was positioned as an âacceptable alternativeâ. These recommendations were endorsed by the joint European Society of Cardiology/American College of Cardiology/American Heart Association/World Heart Federation task force for the definition of myocardial infarction.
Cardiac troponin âthe best single markerâ
Levels of cardiac troponin are dependent on infarct size, and directly indicate the prognosis following MI. Indeed, in recent years, some experts suggest that CTnI and CTnT have âdisplaced myoglobin and creatine kinase-MB as the preferred markers of myocardial injury.â
In 2013, a Health Technology Asssessment (HTA) by Britainâs National Institute for Health Research (NIHR) concurred with this view, observing that âhigh-sensitivity cardiac troponin is the best single marker in patients presenting with chest pain.â Additional measurements of myoglobin or creatine kinase-MB, it noted were ânot clinically effective or cost-effective.â
Debate on troponin not over
Nevertheless, considerable debate remains about the utility of troponin in real world CVD management. Although patients with undetectable troponins are considered to have excellent short-term prognosis, levels may be undetectable âfor six hours after the onset of myocardial cell injury,â making myoglobin âa preferred early markerâ for MI. This limitation, which seems to go against the 2013 NIHR Health Technology Assessment, is also acknowledged by some proponents of troponin, who admit that although it âmay be useful for risk assessment and managementâ in asymptomatic populations, there is no evidence that it confers âan advantage in the context of MI diagnosis.â In addition, they also note that âcTnI assays are not standardized; thus, there can be a substantial difference in values depending on the assay used.â
Defining assay sensitivity, differentiating troponin I and T
One challenge lies in the definition of a âhigh sensitivityâ assay, which can measure cTn in the single digit range of nanograms per litre. The term is used by vendors âfor marketing purposes,â and there âis still no consensusâ regarding its application. Making matters tougher is the fact that most manufacturersâ claims for assay precision âcannot be achieved in clinical laboratories.â
In effect, the jury on troponin is likely to be out for some time to come, accompanied by continuing uncertainties.
For instance, Britainâs respected health advisory site patient.co.uk suggests that troponin I and T âare of equal clinical valueâ while a 2010 guideline from NICE (National Institute for Healthcare and Clinical Excellence) advises taking a blood sample for troponin I or T as âpreferred biochemical markers to diagnose acute MI.â
However, a very recent study published by the Journal of the American College of Cardiology finds that patients with neuromuscular disease can show elevated levels of troponin T but not I, thus questioning the guidelines which regard both as being âequally sensitive and specific for the diagnosis of myocardial injury.â
These may be some of the reasons why the US Food and Drug Administration (FDA) decided in June 2014 to discuss clarification of claims and protocols with vendors of troponin assays, in order to âmodernize the performance evaluation and regulatory review.â In Britain, NICE is currently updating its 2010 guideline.
The role of B-type natriuretic peptide
Once acute MI is confirmed, a variety of other biomarkers are used to help make assessments.
One of the most promising of these is B-type natriuretic peptide or BNP, designated by the FDA in the year 2000 as a Class II diagnostic device.
Nevertheless, it is important to underline that only troponin has been used to direct therapeutic intervention. Though it is evident that the adoption of proven new biomarkers will increase prognostic accuracy, they have yet to be tested to alter outcomes of therapeutic intervention.
Thus, in spite of statements from reputable sources claiming that BNP is âalready used to diagnose heart failure,â the truth is somewhat different, with the difference in the details. At the end of 2013, the US Agency for Healthcare Research and Quality (AHRQ), investigated BNP and the related N-terminal proBNP (NT-proBNP) for detecting heart failure (HF). The findings were guarded: âBNP and NT-proBNP had good diagnostic performance for ruling out HF but were less accurate for ruling in HF.â In addition, it found that the âtherapeutic value was inconclusive.â
Other biomarkers remain valuable
In the meanwhile, clinicians in emergency settings have recourse to a variety of other established CVD risk markers, such as cholesterol. âResearch is also under way on markers with strong predictive value that are not used in the clinic for cardiovascular disease risk prediction, such as fibrinogen, vitamin D, and cystatin C.â Some of these âare of special interest as these may prove to be valuable biomarkers in the future.â
To have clinical utility, however, such biomarkers will need to provide risk assessments independently of other established markers. They also require the presence of standardized assays which are specific and sensitive for the markers, with easy-to-interpret results.
In effect, biomarker-mediated approaches to CVD need to yield superior patient outcomes compared to current standard-of-care management schemes.
Whilst in many less developed countries there is a paucity of diagnostic testing and appropriate therapies, we in the West are suffering from the âmodern epidemicâ of over-diagnosis and over-treatment. Todayâs highly sensitive biomarker and imaging tests increasingly identify asymptomatic or very mild conditions that if left untreated would not cause symptoms or reduce longevity. A recent report on mammography screening in the UK suggested that 19% of breast cancers were over-diagnosed, and a US task force concluded that PSA-based prostate cancer screening over-diagnosed up to 50% of tumours. Other over-diagnosed and over-treated conditions include thyroid cancers as well as a range of cardiovascular diseases, chronic kidney disease and ADHD. At best treating such subjects is an imprudent use of health service funds; at worst âpatientsâ suffer both psychological and physical harm from their diagnosis and subsequent treatment. Of course effective screening for cancer and other serious conditions is vital, but how can the problem of over-diagnosis be at least alleviated when tests (and cut-off values) must be sensitive enough to detect pathologies that really require treatment?
When diagnostic tests are evaluated for accuracy the average sensitivity and specificity are reported. But of course individuals vary, and diseases have stages of severity. What is needed is the identification of those patients for whom treatment will do more good than harm. Similarly average results in therapeutic trials may be positive, so negative effects in some patient groups are not evident, but again the potential benefit of a treatment should be weighed against possible harm according to disease severity. And subjects being screened should surely be informed about the risk of over-diagnosis. Yet in a recent random sample of 500 Australians, only 10% of the women who had had mammography, and 18% of the men who had had prostate cancer screening reported that they had been told about the limitations of these tests.
There is also an urgent need to scrutinize the panels of medical professionals setting disease definitions. Diagnostic thresholds are frequently lowered without considering the balance between good and harm of treating the additional patient group who have a lower risk or milder symptoms. And although it may sound cynical, panels with three quarters of the members having multiple ties to pharmaceutical companies â some of which will directly benefit from an increased number of patients with the disease under discussion â surely canât be unbiased!
Hopefully appropriate action can be taken before the seemingly inexorable trend towards over-diagnosis makes patients of us all!
November 2024
The leading international magazine for Clinical laboratory Equipment for everyone in the Vitro diagnostics
Beukenlaan 137
5616 VD Eindhoven
The Netherlands
+31 85064 55 82
info@clinlabint.com
PanGlobal Media is not responsible for any error or omission that might occur in the electronic display of product or company data.
This site uses cookies. By continuing to browse the site, you are agreeing to our use of cookies.
Accept settingsHide notification onlyCookie settingsWe may ask you to place cookies on your device. We use cookies to let us know when you visit our websites, how you interact with us, to enrich your user experience and to customise your relationship with our website.
Click on the different sections for more information. You can also change some of your preferences. Please note that blocking some types of cookies may affect your experience on our websites and the services we can provide.
These cookies are strictly necessary to provide you with services available through our website and to use some of its features.
Because these cookies are strictly necessary to provide the website, refusing them will affect the functioning of our site. You can always block or delete cookies by changing your browser settings and block all cookies on this website forcibly. But this will always ask you to accept/refuse cookies when you visit our site again.
We fully respect if you want to refuse cookies, but to avoid asking you each time again to kindly allow us to store a cookie for that purpose. You are always free to unsubscribe or other cookies to get a better experience. If you refuse cookies, we will delete all cookies set in our domain.
We provide you with a list of cookies stored on your computer in our domain, so that you can check what we have stored. For security reasons, we cannot display or modify cookies from other domains. You can check these in your browser's security settings.
.These cookies collect information that is used in aggregate form to help us understand how our website is used or how effective our marketing campaigns are, or to help us customise our website and application for you to improve your experience.
If you do not want us to track your visit to our site, you can disable this in your browser here:
.
We also use various external services such as Google Webfonts, Google Maps and external video providers. Since these providers may collect personal data such as your IP address, you can block them here. Please note that this may significantly reduce the functionality and appearance of our site. Changes will only be effective once you reload the page
Google Webfont Settings:
Google Maps Settings:
Google reCaptcha settings:
Vimeo and Youtube videos embedding:
.U kunt meer lezen over onze cookies en privacy-instellingen op onze Privacybeleid-pagina.
Privacy policy