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.
Diagnosis of infection with the parasitic roundworm Strongyloides stercoralis is currently done by stool sample culture to detect active larvae. However, the sensitivity of this method can be as low as 28%. This article describes how cell-free parasite DNA can be detected in urine when the results of stool-sample testing are negative.
by Dr Clive Shiff and Dr Alejandro Krolewiecki
Introduction
Of the neglected tropical diseases, Strongyloides stercoralis infection has emerged as a global problem because it is difficult to diagnose and is often silent but long-lived [1]. Currently the definitive test is performed by examining or making a culture of fresh stool to detect active larvae. Serological analysis for specific antibodies is also used but this is far from definitive [2]. This intestinal parasite has an unusual life cycle which is still somewhat enigmatic and can have dire implications if the patient becomes immunosuppressed [3]. This can happen as a patient ages but also if, for some reason, is placed on immunosuppression therapy. To put this in perspective it is important to appreciate the complexity of this infection and the importance of a simple effective diagnostic test.
The infection is caused by parthenogenetic females that live in the upper reaches of the intestinal tract. Unlike hookworms, which also have adults in the gut, Str. stercoralis does not lay eggs which exit in the stool, embryonate and hatch outside the body, Str. stercoralis females incubate the eggs and deposit the eggs in the intestinal mucosa from which the first-stage rhabditiform larvae emerge in large numbers. Some larvae pass in the stool moult, and commence a free-living sexual phase with male and female adults living in the human fecal material. Free-living stages are not parasitic but after several cycles the larvae change from producing the benign, rhabditiform stage to a parasitic filariform stage. These are infective and will penetrate the skin of anyone approaching or contacting the fecal mass. The parasitic stage occurs after the second moult producing the stage called ‘L3’. These larvae secrete proteolytic enzymes and are tissue invasive. However, not all the larvae produced by the parthenogenetic gut parasites are voided in the stool. A proportion of these larvae moult internally and commence the autoinfection stage. They reinvade the host mucosa and reinfect the host and are distributed round the body in the blood and other fluids. In immunocompetent persons these larvae are killed off and their matter is finally excreted through the urine [3]. In people who become immunosuppressed, these larvae continue to survive and accumulate in large numbers and constitute an urgent, life-threatening condition.
Detecting species-specific DNA from urine
Cell-free DNA of parasite origin has been detected in urine of patients with a number of blood-borne and tissue-dwelling parasites. This has been shown with malaria [4], urogenital schistosomes [5], Schistosoma mansoni [6] and others. In all these publications detection of DNA from urine was the most sensitive of serology, parasitological examination of excreta or antigen capture test and the specificity was equal to detection of eggs in excreta [7]. There is also an advantage in using urine specimens. It is simple and can be collected almost on demand. For this work the specimen is filtered through a standard filter paper cone. Approximately 40 ml of urine is filtered, and then the paper is removed from the beaker, opened and allowed to dry in a fly-proof, clean area [8]. When dry each filter is placed in a sealable zip lock plastic bag with a small desiccant capsule. Papers can be stored at 4 °C for months without deterioration of the parasite DNA. In the field when survey work is carried out, urine collection can be carried out simply and in a single day, but filtration and drying of the filters needs to be done within 3 to 4 hours of collection as DNA is degraded by long storage in the urine specimen.
Methods
Ethical clearances
The specimens were collected as part of an ongoing programme to find and cure infections of soil-transmitted helminths by the Ministry of Health and approved by Commité de Ética Colegio Médico de Salta, Salta, Argentina and Johns Hopkins University (IRB number 6199).
Extraction of parasite DNA from filter paper
Filter papers (Whatman No. 3, 12.5 cm diameter) clearly labelled with pencil received in the laboratory are processed as follows. Using a metal punch fifteen 1.00 mm discs are removed from the apex of the quadrant sampled. These are placed in a sterile 1.5 mL Eppendorf tube and 600 µL of nuclease free water added, then incubated at 95 °C for 10 min, and subject to gentle agitation overnight at room temperature. Tubes were then centrifuged at 4000 r.p.m. for 5 min and the supernatant was removed and processed for DNA extraction. We used QIAmpDNA Blood Mini Kit (Qiagen) according to manufacturer’s protocol. The amount of recovered DNA was measured by NanoDrop, ND-1000 spectrophotometer (Thermo Scientific) and stored at −20 °C [9].
Identification of specific Str. stercoralis DNA fragment
Previous work [10] has shown that tandem repeat DNA composed a high proportion of genomic DNA, and these repeats incorporate smaller repeat fragments of DNA. Small fragments of parasite-specific DNA, are found nested within tandem repeats. GenBank AY028262 is such a fragment. Primers for a 125-bp fragment were designed using PrimerQuest Tool (IDT) these are:
Forward (SSC-F) 5´-CTC AGC TCC AGT AAA GCA ACA G-3´
Reverse (SSC-R) 5´-AGC TGA ATC TGG AGA GTG AAG A-3´.
The sequence amplified by these primers was compared with a Blast search against total GenBank data and found only to amplify Str. stercoralis DNA. They were also tested against DNA from three Ancylostoma spp., Sch. mansoni and Sch. haematobium and found only to amplify a product from Str. stercoralis [9].
Amplification and visualization
PCR amplification in 15-µL volume with 2× Taq Mastermix (New England Biolabs), 0.75 µL of 10 µM of each primer, 1–2 µL (20–100 ng/µL) of product DNA made to volume with PCR-grade water (Sigma-Aldrich). The protocol, denaturation at 95 °C to 10 min and 35 cycles at 95 C for 1 min, 63 °C for 1 min 30s, 72 °C for 1 min and a final extension at 72 °C for 10 min. To confirm amplicon size products, were resolved on a 2% agarose gel and stained with Ethidium Bromide (Sigma-Aldrich) [9].
Results
Limits of detection
Genomic DNA from Str. stercoralis was diluted and titrated sequentially in concentration from 2 ng/µL to 2 fg/µL to determine the extinction level under standard amplification procedure. Amplifications were performed in duplicate to ensure reproducibility. Products amplified were cleaned with ExoSAP-IT (Afflymetrix Inc.), sequenced and compared with the Str. stercoralis repeat sequence in GenBank (AY028262) to ensure confirmation. In Figure 1 the limit of detection was 20 pg of target DNA.
Diagnostic efficacy
A study was conducted to compare the diagnostic efficacy of parasitological copro-diagnostic methods with DNA detection. For this specimens of stool and urine were collected from 125 individuals living in endemic regions of northern Argentina. The stool specimens were examined fresh using three parasitological tests, concentration- sedimentation, Harada-Mori and Baermann culture methods. Urine samples were filtered as outlined above, dried and sent to the laboratory at Johns Hopkins for DNA extraction and amplification.
The results are given in Table 1 comparing the results of stool versus urine analysis. The prevalence when stool only, 28% is compared with the DNA detection 44.8%, the difference in prevalence is a highly significant 62% difference (P=0.0058). With further analysis comparing the two procedures in the same community, detection of DNA in the urine is more sensitive with significant difference again, 87.5% (95% CL 76.8–94.4) against 56.5% (CL 42.3–69.0%). Specificity in both tests was 100%.
Discussion
There are important reasons for the development of highly sensitive and specific diagnostic tests for the neglected tropical diseases. These relate to modern attempts to limit or eliminate these diseases from much of the endemic areas [11]. However, most parasitic infections have been sustained in their communities for evolutionary time and the parasites have adapted effectively to sustain their populations. This has resulted in very high replicative stages in the life cycle, for instance schistosomes produce large and sustained numbers of cercariae in the snail intermediate hosts from a single miracidium [12]. With strongyloidiasis the multiplication occurs in the host through effective autoinfection; hence, effective control must identify all cases to eliminate the condition. In this work the difference between a prevalence of 28% and 44.8%, means missing almost half of the population at risk. Serological diagnoses are available, but authorities are not satisfied with either the specificity or sensitivity of these tests [13].
The work described there opens an avenue to help ameliorate these problems on two counts. First there is an improvement in sensitivity without loss of specificity, albeit the process requires the use of DNA amplification and detection equipment. This has been mentioned in numerous review articles, but in reality it is an excuse rather than a reason because there are few countries in the world now where there is no access to such equipment. Furthermore the ‘loop mediated amplification procedure’ (LAMP) has been applied to most of these diagnostic methods with success, so amplification is not a real problem. The main difficulty has been in collecting and storing specimens. This has been solved with the use of urine as a vehicle for diagnostic DNA. There are two reasons. First, urine can be obtained on demand; there is no need for a long wait. Second, the specimen is easily collected: the procedure is non-invasive and with simple equipment the sample can be filtered through standard Whatman No. 3 filter paper within minutes of collection. The collection of urine samples for DNA testing has already been done in Nigeria [8] and elsewhere, where colleagues have implemented the work.
Several laboratories are focusing on stool collections as so many soil-transmitted helminths are transmitted by feces. In a hospital environment, collection of a stool sample is a straightforward procedure that can be carried out under clean and safe-handling conditions. DNA detection can be carried out on preserved feces, and using real-time PCR multiplex procedures DNA from various sources (parasitic) can be identified from a single sample and the procedure is currently in use [14]. Although there is an advantage in multiple diagnoses from a single stool, the sensitivity will depend on whether there are actual organisms in the stool examined. In low-density infections, there are times when there is no parasite material in the feces, which will give a false negative response [15]. It has been shown with Sch. mansoni infections, DNA was detected in urine when there were no eggs of the parasite seen in stool [6].
Conclusions
Although this method may not be feasible for all soil-transmitted helminths, detection of parasite-specific DNA in urine seems the best way of achieving optimum sensitivity. The use of urine also has added advantages over stool collection, primarily because it is available more or less on demand, it is simple to handle, does not require fume extraction hoods, it is not dangerous to handle and can be processed in the field, and once collected on dry filter paper it is easily and economically transported.
References
1. Schad G. Morphology and life history of Strongyloides stercoralis. In: Gove DI (Ed) Strongyloidiasis: a major roundworm infection of man. Taylor and Francis 1989.
2. Krolewiecki AJ, Ramanathan R, Fink V, McAuliffe I, Cajal SP, Won K, Juarez M, Di Paolo A, Tapia L, et al. Improved diagnosis of Strongyloides stercoralis using recombinant antigen-based serologies in a community-wide study in northern Argentina. Clin Vaccine Immunol 2010; 17(10): 1624–1630.
3. Schad G, Aikens L, Smith G. Strongyloides stercoralis: is there a canonical migratory route through the host? J Parasitol 1989; 75: 740–749.
4. Mharakurwa S, Simoloka C, Thuma PE, Shiff CJ, Sullivan DJ. PCR detection of Plasmodium falciparum in human urine and saliva samples. Malar J 2006; 5: 103.
5. Ibironke OA, Phillips AE, Garba A, Lamine SM, Shiff C. Diagnosis of Schistosoma haematobium by detection of specific DNA fragments from filtered urine samples. Am J Trop Med Hyg 2011; 84(6): 998–1001.
6. Lodh N, Mwansa JC, Mutengo MM, Shiff CJ. Diagnosis of Schistosoma mansoni without the stool: comparison of three diagnostic tests to detect Schistosoma mansoni infection from filtered urine in Zambia. Am J Trop Med Hyg 2013 July; 89(1): 46–50.
7. Krolewiecki AJ, Lammie P, Jacobson J, Gabrielli AF, Levecke B, Socias E, Arias LM, Sosa N, Abraham D, et al. A public health response against Strongyloides stercoralis: time to look at soil-transmitted helminthiasis in full. PLoS Negl Trop Dis 2013; 7(5): e2165.
8. Ibironke OA, Koukounari A, Asaolu S, Moustaki I, Shiff C. Validation of a new test for Schistosoma haematobium based on detection of the Dra1 DNA repeat fragment in urine: evaluation through latent class analysis. PLoS Negl Trop Dis 2012; 6(1): e1464.
9. Lodh N, Caro N, Sofer S, Scott A, Krolewiecki A, Shiff C. Diagnosis of Strongyloides stercoralis: detection of parasite-derived DNA in urine. Acta Tropica 2016; 163: 9–13.
10. Hamburger J, Turetski T, Kapeller I, Deresiewicz R. Highly repeated short DNA sequences in the genome of Schistosoma mansoni recognized by a species-specific probe. Mol Biochem Parasitol 1991; 44: 73–80.
11. Lo NC, Addiss DG, Hotez PJ, King CH, Stothard R, Evans DS, Colley DG, Lin W, Coulibaly JT, et al. A call to strengthen the global strategy against schistosomiasis and soil-transmitted helminthiasis: the time is now. Lancet Infect Dis 2016.
12. Shiff C. The importance of definitive diagnosis in chronic schistosomiasis, with reference to Schistosoma haematobium. J Parasitol Res 2012; 2012: 761269.
13. Bisoffi Z, Buonfrate D, Sequi M, Mejia R, Cimino RO, Krolewiecki AJ, Albonico M, Gobbo M, Bonafini S, et al. Diagnostic accuracy of five serologic tests for Strongyloides stercoralis infection. PLoS Negl Trop Dis 2014; 8(1): e2640.
14. Basuni M, Muhi J, Othman N, Verweij JJ, Ahmad M, Miswan N, Rahumatullah A, Aziz FA, Zainudin NS, Noordin R. A pentaplex real-time polymerase chain reaction assay for detection of four species of soil-transmitted helminths. Am J Trop Med Hyg 2011; 84(2): 338–343.
15. Lodh N, Naples JM, Bosompem KM, Quartey J, Shiff CJ. Detection of parasite-specific DNA in urine sediment obtained by filtration differentiates between single and mixed infections of Schistosoma mansoni and S. haematobium from endemic areas in Ghana. PLoS One 2014; 9: e91144.
The authors
Clive Shiff*1 PhD and Alejandro Krolewiecki2 MD, PhD
1Department of Molecular
Microbiology and Immunology,
Johns Hopkins Bloomberg
School of Public Health,
Baltimore, MD 21205, USA
2Instituto de Investigaciones en
Enfermedades Tropicales,
San Ramón de la Nueva Orán 4530,
Salta, Argentina
*Corresponding author
E-mail: cshiff1@jhu.edu
by Carrie A. Chadwick The relationship between anti-glycolipid antibodies and peripheral neuropathies has for many years been studied by laboratory investigations because of the antibody-phenotype associations identified. This has led to the discovery of a number of peripheral nerve conditions affected by the presence of anti-glycolipid antibodies. This article describes some of the conditions associated […]
MILFORD, Mass.–(BUSINESS WIRE)–Waters Corporation (NYSE:WAT) announced today that its fully-validated Waters® MassTrak™ Vitamin D Solution is now CE-marked in accordance with IVD Directive 98/79/EC for the quantitative measurement of 25(OH) D2 and D3 (25-OH-VitD) from human plasma and serum. The IVD LC-MS/MS system solution is a complete solution consisting of reagents and consumables, instrumentation, and software and support services for clinical laboratories routinely measuring vitamin D in patient samples*. The Vitamin D solution is currently only available for sale in Europe.
“Our MassTrak Vitamin D Solution is the first, CE-marked, single-vendor system to measure both vitamins 25(OH) D2 and D3 independently and accurately from human plasma and serum in a single analysis,” said Jose Castro-Perez, Ph.D., Director of Health Sciences, Global Marketing Operations, Waters Corporation. “It’s the ideal solution for clinical laboratories performing (25-OH-VitD) analysis on a routine basis and which may want to standardize (25-OH-VitD) analysis on a single-vendor platform.”
Vitamin D, the main source of which is from dietary intake, is part of a family of fat-soluble secosteroids and has two major forms – ergocalciferol (plant derived Vitamin D2) and cholecalciferol (animal derived Vitamin D3). Vitamin D is also derived from the conversion of 7-dihydrocholesterol to Vitamin D3 following exposure of skin to sunlight. Increasingly, research is revealing the importance of vitamin D in protecting against a host of health problems. In the absence of vitamin D, bones can become brittle and thin. The research also suggests that vitamin D plays a role in modulating cell growth, the function of neuromuscular and immune systems, and reducing inflammation. It also indicates that those most at risk of a vitamin D deficiency are those who shun the sun, have milk allergies, adhere to a strict vegan diet, or suffer from certain diseases.
Due, in part, to mounting evidence of the importance of vitamin D to human health and to news media coverage, Medicare reimbursement volumes of vitamin D laboratory tests rose 83-fold from 2000 – 20101.
The MassTrak Vitamin D Solution is configured with the Waters ACQUITY UPLC® I-Class/ Xevo® TQD IVD System, the MassTrak Vitamin D Kit and MassLynx® (IVD) Mass Spectrometry Software. The MassTrak Vitamin D Kit provides metrological traceability to the National Institute of Standards and Technology (NIST) Standard Reference Material (SRM) 2972, enabling standardization and lot- to-lot consistency for an easy comparison of results from multiple laboratories. For high-volume vitamin D testing, the system is designed for use with an automated liquid handler and custom pipetting script provided with the kit.
LC-MS/MS in the Clinical Laboratory
In pursuit of better accuracy and precision for clinical laboratory analyses, liquid chromatography tandem mass spectrometry (LC-MS/MS) is rapidly becoming the analytical technique of choice. Waters IVD LC-MS/MS medical devices provide clinical laboratories access to technologies delivering selectivity, sensitivity and versatility – performance characteristics that traditional methods, such as immunoassays, often struggle to meet.
Designed to optimize workflow from pre-analytics to post-analytics, our system portfolio offers a wide variety of options for the routine use of LC-MS/MS in the clinical laboratory.
Advances in circulating biomarker research have led to the use of blood samples to characterize cancer patients’ tumour DNA where a lack of tumour tissue prevents molecular testing. This is critical for non-small cell lung cancer (NSCLC) patients who require tumour molecular characterization in order to access life-extending treatments that would be denied without biopsy. Here we describe a new liquid biopsy diagnostic service for NSCLC patients at the All Wales Medical Genetics Service, Cardiff, UK.
by Dr Angharad Williams, Dr Daniel Nelmes, Helen Roberts and Dr Rachel Butler
Liquid biopsies and cell-free circulating tumour DNA (ctDNA) in clinical practice
The term ‘liquid biopsy’ comes from the sampling of a cancer patient’s tumour DNA from a simple, non-invasive blood test rather than an invasive surgical biopsy. This circulating tumour DNA (ctDNA) is a small fraction of the total cell-free circulating DNA (cfDNA) and consists of short strands of DNA shed by degrading tumour cells directly into a patient’s bloodstream. The levels of ctDNA present will vary greatly based on clinical factors such as proximity of sampling to chemotherapy or radiotherapy, as well as the burden and activity of the tumour [1].
Genetic mutations within the patient’s tumour are detectable at extremely low levels in the ctDNA in the blood [2]. The detection of such mutations provides many potential uses for ctDNA as a biomarker in disease diagnosis and screening, monitoring of therapy response and resistance and detection of minimal residual disease and relapse [3–6].
The many advantages for using ctDNA as a biomarker rest on the fact that ctDNA can be simply extracted from blood; therefore, invasive biopsy procedures can be avoided. Such simple blood sampling is beneficial if the patient is too ill for invasive surgery and is also useful if biopsy-based tumour analysis has failed; thus, unnecessary re-biopsies can be averted. Another benefit of the use of ctDNA over biopsies is that serial blood samples can be taken to replace the need for a re-biopsy to monitor a patient’s response to therapy in ‘real-time’ in the clinic. Practically, blood samples can be arranged, taken and sent for processing at a much faster pace than surgery, gaining valuable time for patients who are in need of urgent cancer-related treatments.
There are, however, potential pitfalls in using ctDNA as a diagnostic biomarker that should be considered prior to setting up a ctDNA-based diagnostic service as well as when interpreting genetic results from ctDNA (summarized in Figure 1). The greatest concern is the fragile nature of cfDNA molecules [7], which means that cfDNA will degrade in a blood sample to undetectable levels the longer that the blood is left unprocessed. Efficient centrifugation and separation of the blood to plasma and storage at −80 °C can be used to halt degradation of cfDNA. In cases where analysis of the cfDNA sample identifies no genetic mutations, this raises the important question of whether the patient was actually shedding ctDNA at the time of the blood sampling or did the ctDNA degrade prior to sample processing? This indicates the unfortunate possibility of false negative results when using ctDNA in the diagnostic setting. Another important factor to consider is that the level of a mutation in the ctDNA, which can quite often be as low as ≤1% mutated ctDNA to wild-type patient cfDNA [8]. Thus, only highly sensitive molecular analysis options should be considered for diagnostic testing strategies using ctDNA.
Molecular analysis of the epidermal growth factor receptor gene in non-small cell lung cancer patients
The epidermal growth factor receptor gene (EGFR) encodes the EGRF protein, a signalling protein that is part of the cellular pathways that control normal cell growth, differentiation and angiogenesis [9]. Approximately 10–20% of ethnically Caucasian non-small cell lung cancer (NSCLC) patients with the adenocarcinoma histological subtype will have a DNA mutation in the EGFR gene, which will activate abnormal constitutive signalling and tumorigenesis [10].
The most common sensitizing EGFR mutations, which represent 85% of known activating EGFR mutations in NSCLC, are the exon 21 point mutation c.2573T>G (p.Leu858Arg) and in-frame deletions in exon 19 [9]. These activating mutations provide a convenient target for first and second generation tyrosine kinase inhibitor (TKI) treatments such as gefitinib (Iressa®, AstraZeneca) [11–13] and act as positive predictive biomarkers for response to these drugs. Traditionally, for patients to access these TKI treatments, tumour biopsy in the form of a formalin-fixed sample is tested for evidence of these activating EGFR mutations at clinical genetic testing centres, such as the All Wales Medical Genetics Service (AWMGS) in Cardiff. However, preservation of the tumour biopsy as formalin-fixed paraffin-embedded (FFPE) tissue leads to a number of issues with genetic analysis including poor quality and yields of DNA (noted in Figure 1). Additionally, a large proportion of NSCLC patients are not well enough to have a biopsy taken and so genetic analysis of tumour DNA and subsequent access to TKI treatments is not possible. This inequity in service provision indicated a clinical need to expand current testing options for NSCLC patients to reach those patients who cannot access TKI-based stratified medicine treatment options. To address this clinical need, a ctDNA-based diagnostic NHS service was developed within AWMGS to detect activating EGFR mutations from patient blood samples in order to alleviate the need for biopsy.
In addition to the availability of first and second generation TKIs, a new third generation TKI, osimertinib (Tagrisso®, AstraZeneca), has recently been made available to a specific group of NSCLC patients. Approximately 50% of patients on first and second generation TKIs will develop an EGFR resistance mutation, c.2369C>T (p.Thr790Met) (commonly known as T790M), leading to disease progression [6]. Since October 2016, osimertinib (Tagrisso®, AstraZeneca), has been available to UK patients shown to harbour the T790M mutant in their tumour via either biopsy or ctDNA analysis through the NHS Cancer Drugs Fund [14]. CtDNA testing has become a popular method of testing for resistance mutations as it mitigates the need for a second invasive biopsy for the patient and, also, serial blood samples can be used to track the patient’s response over a period of time [15].
Establishing the ctDNA-based NSCLC stratified medicine service in the All Wales Medical Genetics Service
Since 2009, the AWMGS has been providing stratified medicine services for NSCLC patients, as well as metastatic colorectal cancer patients, melanoma and gastrointestinal stromal tumour patients in Wales. Though all of these services are based on FFPE tumour analysis, we have developed a wealth of experience in using ctDNA from blood in the field of clinical trials. By 2015, following a number of successful ctDNA-based feasibility studies by laboratory staff and research students, we were confident that we had the knowledge and expertise to bring ctDNA into service, and were one of the first laboratories in the UK to do so.
Owing to the inherent shortcomings of using ctDNA as a biomarker, discussed previously, the following questions were deliberated during validation to find the most appropriate testing methods for the diagnostic service:
To guarantee sample quality and maintain sufficient levels of ctDNA, we have imposed stringent quality measures on the blood collection and dispatch. The main requirement is that blood samples should be taken in a specialist preservative tube such as CellSave Preservative Tubes (Janssen Diagnostics) or Cell-Free DNA BCT® (Streck) and must reach the laboratory for processing within a strict 96-hour window. This was decided on after discussion with other research groups and internal investigations on the stability of ctDNA in blood and the use of preservative tubes [7].
The sensitivity of the molecular ctDNA assay was paramount in our decision to use the recently developed technology droplet digital polymerase chain reaction (ddPCR) by Bio-Rad (Bio-Rad Laboratories, Inc, California, USA). ddPCR is a highly sensitive fluorescence-based PCR method with an extreme lower limit of detection of 0.0001% of mutant DNA in a wild-type background, which makes it the superior choice over other technologies such as next-generation sequencing and quantitative PCR (qPCR). Practically, in the service, we have detected EGFR mutations in patient ctDNA at an abundance as low as 0.7%.
The AWMGS now provides ctDNA-based testing of the EGFR gene in NSCLC patients at both first-line testing for sensitizing mutations and for resistance mutation testing on patient progression on TKIs (Figure 2). The service launched across Wales in April 2016 and has since been expanded to provide testing for certain centres in the South West of England with funding from AstraZeneca. A year on, over 100 patients have been tested, the majority (approximately 60%) of patient referrals have been for T790M progression testing to avoid repeat biopsies for patients. Six patients, for whom TKIs were previously inaccessible due to failed FFPE-based testing or inability to biopsy, were successfully tested through the ctDNA service and are now receiving first-line EGFR TKI therapy following the detection of activating EGFR mutations in ctDNA.
Ongoing and future developments
The field of liquid biopsies is steadily gaining pace in the UK and abroad with a number of centres now providing EGFR ctDNA testing. New circulating biomarkers, such as exosomes and circulating tumour cells, are coming through from translation research and have vast potential in the field of stratified medicine. At the AWMGS, we aim to expand our current liquid biopsy testing in the near future with targets for both metastatic colorectal cancer and metastatic melanoma.
References
1. Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer 2011; 11: 426–437.
2. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014; 6: 224ra24.
3. Chen KZ, Lou F, Yang F, Zhang JB, Ye H, Chen W, Guan T, Zhao MY, Su XX, et al. Circulating tumor DNA detection in early-stage non-small cell lung cancer patients by targeted sequencing. Sci Rep 2016; 6: 31985.
4. Dawson S-J, Tsui DWY, Murtaza M, Biggs H, Rueda OM, Chin SF, Dunning MJ, Gale D, Forshew T, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med 2013; 368: 1199–1209.
5. Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DW, Kaper F, Dawson SJ, Piskorz AM, Jimenez-Linan M, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 2012; 4: 136ra68.
6. Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, Parkinson C, Chin SF, Kingsbury Z, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013; 497: 108–112.
7. Rothwell DG, Smith N, Morris D, Leong HS, Li Y, Hollebecque A, Ayub M, Carter L, Antonello J, et al. Genetic profiling of tumours using both circulating free DNA and circulating tumour cells isolated from the same preserved whole blood sample. Mol Oncol 2016; 10: 566–574.
8. Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem 2015; 61: 112–123.
9. Jänne PA, Engelman JA, and Johnson BE. Epidermal growth factor receptor mutations in non–small-cell lung cancer: implications for treatment and tumor biology. J Clin Onc 2005; 23: 3227–3234.
10. Li T, Kung H-J, Mack PC, Gandara DR. Genotyping and genomic profiling of non-small-cell lung cancer: implications for current and future therapies. J Clin Onc 2013; 31: 1039–1049.
11. Douillard JY, Ostoros G, Cobo M, Ciuleanu T, Cole R, McWalter G, Walker J, Dearden S, Webster A, et al. Gefitinib treatment in EGFR mutated Caucasian NSCLC: circulating-free tumor DNA as a surrogate for determination of EGFR status. J Thorac Oncol 2014; 9: 1345–1353.
12. Goto K, Ichinose Y, Ohe Y, Yamamoto N, Negoro S, Nishio K, Itoh Y, Jiang H, Duffield E, et al. Epidermal growth factor receptor mutation status in circulating free DNA in serum: from IPASS, a phase III study of gefitinib or carboplatin/paclitaxel in non-small cell lung cancer. J Thorac Oncol 2012; 7: 115–121.
13. National Institute for Health and Care Excellence (NICE). Gefitinib for the first-line treatment of locally advanced or metastatic non-small-cell lung cancer. Technology appraisal guidance [TA192] 2010. [https: //www.nice.org.uk/guidance/ta192]
14. NICE. Osimertinib for treating locally advanced or metastatic EGFR T790M mutation-positive non-small-cell lung cancer. Final appraisal determination [TA10022] 2016. [https: //www.nice.org.uk/guidance/GID-TA10022/documents/final-appraisal-determination-document]
15. Sundaresan TK, Sequist LV, Heymach JV, Riely GJ, Jänne PA, Koch WH, Sullivan JP, Fox DB, Maher R, et al. Detection of T790M, the acquired resistance EGFR mutation, by tumor biopsy versus noninvasive blood-based analyses. Clin Cancer Res 2016; 22: 1103–1110.
The authors
Angharad Williams1 PhD, Daniel Nelmes2,3 PhD, Helen Roberts1 BSc and Rachel Butler*1 FRCPath
1All Wales Medical Genetics Service,
NHS Wales, The Institute of Medical
Genetics, Cardiff and Vale University LHB,
University Hospital of Wales, Cardiff
CF14 4XW, Wales, UK
2School of Medicine, Cardiff University, Cardiff CF14 4XN, Wales, UK
3Velindre Cancer Centre, Cardiff
CF14 2TL, Wales, UK
*Corresponding author
E-mail: Rachel.Butler@wales.nhs.uk
Leiomyosarcoma and endometrial stromal sarcoma are the most common types of uterine sarcoma, a group of rare and clinically aggressive mesenchymal cancers. These two sarcomas may have overlapping clinical presentation, morphology and protein expression profiles, making their diagnosis occasionally difficult. This article discusses molecular approaches that may be applied to the diagnosis of these two cancers and may generate data expanding our therapeutic options and patient outcome.
by Professor Ben Davidson
Introduction
The majority of cancers affecting the uterine corpus are carcinomas, i.e. tumours of epithelial origin. Uterine sarcomas, tumours that are of mesenchymal origin, are a group of rare and clinically aggressive tumours constituting 7 % of all soft tissue sarcomas and 3 % of malignant uterine tumours [1, 2]. The most common entities within this group are leiomyosarcoma (LMS) and endometrial stromal sarcoma (ESS) [2, 3]. Although LMS and ESS are readily diagnosed based on morphology and a limited immunohistochemistry (IHC) panel in many cases, some tumours may pose diagnostic difficulty, and currently used antibodies are not 100 % sensitive or specific [4]. Improved understanding of the molecular make-up of these tumours may lead to more accurate diagnosis and better understanding of their biology, eventually improving our ability to design targeted therapy approaches with the objective of improving patient outcome.
The genetic make-up of ESS and LMS
Low-grade ESS, the more common type of ESS, is characterized by several gene rearrangements creating fusion genes, of which the first described was fusion of the zinc finger gene 1 JAZF1, located at 7p15, and JJAZ1, also termed SUZ12, at 17q21 through a 7;17-translocation. Other fusions in low-grade ESS include the one between JAZF1 and the PHD finger protein 1 gene (PHF1) in 6p21, as well as between PHF1 and enhancer of polycomb homologue 1 (EPC1) gene at 10p11 and the MYST/Esa1 associated factor 6 gene (MEAF6) at 1p34. X chromosome rearrangements include fusion of the open reading frame CXorf67 and the BCL-6 interacting corepressor (BCOR) gene, both at Xp11, with the MBT domain-containing protein 1 gene (MBTD1) at 17q21 and with the zinc finger CCCH-type containing 7B gene (ZC3H7B) at 22q13, respectively.
High-grade ESS is characterized by a fusion between the tyrosine 3/tryptophan 5 monooxygenase gene (YWHAE) gene at 17p13 and the NUT family member gene (NUTM2; previously known as FAM22) at 10q22, creating YWHAE-NUTM fusion through a 10;17-translocation (reviewed by Davidson and Micci, invited review submitted to Expert Rev Mol Diagn). These alterations were recently confirmed by analysis of the ESS transcriptome and/or whole-exome sequencing, including the application of next generation sequencing [5–7].
The body of data with respect to the molecular characteristics of LMS is more limited. An observation found in several studies is the presence of exon 2 mutations in the mediator complex subunit 12 (MED12) gene on chromosome band Xq13.1 in some LMS. MED12 protein forms complex with MED13, cyclin-dependent kinase 8 (CDK8), and cyclin C, termed the CDK8 submodule of the Mediator, the mediator being a large multiprotein complex regulating transcription [8]. Though less frequent in LMS compared to leiomyomas, the benign counterpart of LMS, this finding appears to be absent in other malignant soft tissue sarcomas, and is rare in carcinomas, and is thus potentially relevant in the diagnostic setting (reviewed by Croce & Chibon [9]).
RNA sequencing of 99 LMS, of which 49 were uterine, identified 3 distinct molecular subtypes. Leiomodin (LMOD1) and ADP-ribosylation factor-like 4C (ARL4C) were found to be markers for type I and II tumours, respectively, and the latter group was associated with poor prognosis when located in the uterus [10].
Comparative molecular analysis of ESS and LMS
Our group performed two studies of uterine LMS and ESS with the aim of identifying novel biomarkers that may expand the arsenal of markers currently used in diagnosing these tumours, as well as improving our understanding of their unique biology.
In the first study, the gene expression profiles of 7 ESS and 13 LMS were compared using the HumanRef-8 BeadChip from Illumina. We identified 549 unique probes that were significantly differentially expressed in the two tumour entities, of which 336 and 213 were overexpressed in ESS and LMS, respectively. Genes found to be overexpressed in ESS included CCND2, ECEL1, ITM2A, NPW, SLC7A10, EFNB3, PLAG1 and GCGR, whereas genes overexpressed in LMS included FABP3, TAGLN, CDKN2A, JPH2, GEM, NAV2 and RAB23. qPCR analysis confirmed these differences for 14 of 16 genes selected for validation. Five protein products were selected for validation by IHC, including the LMS markers fatty acid binding protein (FABP3), transgelin (TAGLN) and neuron navigator 2 (NAV2) and the ESS markers cyclin D2 (CCND2) and integral membrane protein 2A (ITM2A). All were found to be significantly differentially expressed in LMS vs ESS (Fig. 1) [11]. Data for FABP3, TAGLN, NAV2 and CCND2 were recently confirmed in a large (approx. 350 tumours) uterine sarcoma series [Davidson et al., manuscript submitted].
Recently, we compared the microRNA (miRNA) profiles of primary ESS (n=9), primary LMS (n=8) and metastatic LMS (n=8) using Taqman Human miRNA Array Cards. Ninety-four miRNAs were significantly differentially expressed in ESS vs LMS, of which 76 and 18 were overexpressed in ESS and LMS, respectively. Forty-nine miRNAs were differentially expressed in primary and metastatic LMS, among which 45 and 4 were overexpressed in primary and metastatic LMS, respectively. Twenty miRNAs found to be most significantly differentially expressed in primary ESS vs LMS or in primary vs metastatic LMS were further studied in a validation series of 44 tumours using qPCR. Of these, 10 were confirmed to be differentially expressed in these groups, including overexpression of 7 miRNAs (mir-15b, mir-21, mir-23b, mir-25, mir-145, mir-148b and mir-195) in ESS compared to primary LMS. The remaining 3 differentially expressed miRNAs were in comparative analysis of primary and metastatic LMS (lower mir-15a and mir-92a levels and higher mir-31 levels in primary LMS). Differentially expressed miRNA regulated the mitogen-activated protein kinase (MAPK) signaling pathway, Wnt signaling, focal adhesion, the mTOR signaling pathway and the transforming growth factor-β (TGF-β) signaling pathway. As Wnt signaling pathway genes are controlled by miRNAs 15a, 31 and 92a in LMS, we looked at the biological role of Frizzled-6 in LMS cells and found that Frizzled-6 silencing by siRNA significantly inhibited cellular invasion, wound closure and matrix metalloproteinase (MMP-2) activity [12]
Conclusion and future perspectives
Recent years have brought about considerable progress in our understanding of the molecular events occurring in ESS and LMS. Our studies and data from other groups may aid in the diagnosis and classification of these cancers, hopefully providing rationale for targeted therapy. Uterine sarcomas express different cancer-related molecules that may be targeted (reviewed by Cuppens et al. [13]). Anti-hormonal treatment is used in patients with hormone receptor-positive tumours, and expression of progesterone receptor was recently shown to be a prognostic marker in stage I LMS [14]. In two studies, targeting of mTOR, Aurora kinases and other mitotic checkpoint regulators has been suggested as therapeutic modality in LMS [15,16]. Additional studies are likely to identify new relevant targets in the future, hopefully improving the outcome of uterine sarcoma patients.
Acknowledgement
The work of Dr Davidson is supported by the National Sarcoma Foundation at the Norwegian Radium Hospital.
References
1. Toro JR, Travis LB, Wu HJ, Zhu K, Fletcher CD, Devesa SS. Incidence patterns of soft tissue sarcomas, regardless of primary site, in the surveillance, epidemiology and end results program, 1978-2001: an analysis of 26,758 cases. Int J Cancer 2006; 119: 2922–2930.
2. D’Angelo E, Prat J. Uterine sarcomas: a review. Gynecol Oncol. 2010; 116: 131–139.
3. Kurman RJ, Carcangiu ML, Herrington CS, Young RH (Eds.). WHO classification of tumours of female reproductive organs. IARC 2014.
4. Abeler VM, Nenodovic M. Diagnostic immunohistochemistry in uterine sarcomas: a study of 397 cases. Int J Gynecol Pathol. 2011; 30: 236–243.
5. Micci F, Gorunova L, Agostini A, Johannessen LE, Brunetti M, Davidson B, Heim S, Panagopoulos I. Cytogenetic and molecular profile of endometrial stromal sarcoma. Genes Chromosomes Cancer 2016; 55: 834–846.
6. Choi YJ, Jung SH, Kim MS, Baek IP, Rhee JK, Lee SH, Hur SY, Kim TM, Chung YJ, Lee SH. Genomic landscape of endometrial stromal sarcoma of uterus. Oncotarget 2015; 6: 33319–33328.
7. Li X, Anand M, Haimes JD, Manoj N, Berlin AM, Kudlow BA, Nucci MR, Ng TL, Stewart CJ, Lee CH. The application of next-generation sequencing-based molecular diagnostics in endometrial stromal sarcoma. Histopathology 2016; 69: 551–559.
8. Clark AD, Oldenbroek M, Boyer TG. Mediator kinase module and human tumorigenesis. Crit Rev Biochem Mol Biol. 2015; 50: 393–426.
9. Croce S, Chibon F. MED12 and uterine smooth muscle oncogenesis: state of the art and perspectives. Eur J Cancer 2015; 51: 1603–1610.
10. Guo X, Jo VY, Mills AM, Zhu SX, Lee CH, Espinosa I, Nucci MR, Varma S, Forgó E, Hastie T, Anderson S, Ganjoo K, Beck AH, West RB, Fletcher CD, van de Rijn M. Clinically relevant molecular subtypes in leiomyosarcoma. Clin Cancer Res. 2015; 21: 3501–3511.
11. Davidson B, Abeler VM, Hellesylt E, Holth A, Shih IeM, Skeie-Jensen T, Chen L, Yang Y, Wang TL. Gene expression signatures differentiate uterine endometrial stromal sarcoma from leiomyosarcoma. Gynecol Oncol. 2013; 128: 349–355.
12. Ravid Y, Formanski M, Smith Y, Reich R, Davidson B. Uterine leiomyosarcoma and endometrial stromal sarcoma have unique miRNA signatures. Gynecol Oncol. 2016; 140: 512–517.
13. Cuppens T, Tuyaerts S, Amant F. Potential therapeutic targets in uterine sarcomas. Sarcoma 2015; 2015: 243298.
14. Davidson B, Kjæreng ML, Førsund M, Danielsen HE, Kristensen GB, Abeler VM.. Progesterone receptor expression is an independent prognosticator in FIGO stage I uterine leiomyosarcoma. Am J Clin Pathol. 2016; 145: 449–458.
15. Brewer Savannah KJ, Demicco EG, Lusby K, Ghadimi MP, Belousov R, Young E, Zhang Y, Huang KL, Lazar AJ, Hunt KK, Pollock RE, Creighton CJ, Anderson ML, Lev D.. Dual targeting of mTOR and aurora-A kinase for the treatment of uterine leiomyosarcoma. Clin Cancer Res. 2012; 18: 4633–4645.
16. Shan W, Akinfenwa PY, Savannah KB, Kolomeyevskaya N, Laucirica R, Thomas DG, Odunsi K, Creighton CJ, Lev DC, Anderson ML. A small-molecule inhibitor targeting the mitotic spindle checkpoint impairs the growth of uterine leiomyosarcoma. Clin Cancer Res. 2012; 18: 3352–3365.
The author
Ben Davidson1,2 MD, PhD
1Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, N-0310 Oslo, Norway
2University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, N-0316 Oslo, Norway
*Corresponding author
E-mail: bend@medisin.uio.no
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