C286 Figure1 CLI

Proteomics as an alternative diagnostic tool for cervical cancer

Cervical cancer is mainly caused by high-risk human papillomavirus (HPV) infection. The Pap test is the gold standard for early cervical cancer diagnosis. However, the lack of Pap test accessibility accounts for the high rates of cervical cancer mostly in developing regions. Here we discuss recent proteomic approaches towards the development of novel diagnostic and prognostic putative biomarkers.

by Georgia Kontostathi, Dr Jerome Zoidakis, Prof. Nicholas P. Anagnou, Prof. Kalliopi I. Pappa and Dr Manousos Makridakis

Background
Cervical cancer is one of the most common gynecological cancers. It shares many common characteristics such as pathways and regulatory networks with vulvar and endometrial cancer [1]. The majority of cervical cancer incidents are attributed to HPV infection by high-risk oncogenic HPV types (mostly HPV16 and HPV18). HPV infects the basal membrane of cervical epithelium leading to upregulated expression of E6 and E7 oncogenes that cause specific histological lesions such as CIN1 [cervical intraepithelial neoplasia or low-grade squamous intraepithelial lesions (LSIL)], CIN2 and CIN3 [or high-grade squamous intraepithelial lesions (HSIL)] [2, 3].

Cervical cancer is the fourth most common cancer in women worldwide, regarding incidence and mortality. It was responsible for 528 000 incidents of malignancy and 266 000 deaths in 2012, of which more than 85% occurred in developing regions [4]. The high number of cervical cancer cases in developing countries is mainly attributed to the limited availability of diagnostic tools such as Pap smear tests or HPV DNA genotyping that enable detection of early-stage lesions [5].

Current diagnostic methods
The Pap test is the most popular diagnostic technique and is based on the nuclear morphology evaluation of cervical epithelial cells. This test enables the detection of possible lesions at an early stage [6]. However, there is a wide range of sensitivity (from 33.8% to 94.0%) [7, 8] that reflects the main limitation of Pap test: the high inter-observer variability.

Other diagnostic options are based on direct probes, such as Southern blot for HPV genomic analysis. This technique has a relatively low sensitivity, is time-consuming and requires large amounts of purified DNA. More sensitive methods include commercially available kits like Digene’s HC2 test (not HPV type specific), which is based on detection of viral RNA by probes. It is used for patients with minor abnormalities detected by Pap test that need further confirmation. Finally, targeted amplification methods such as PCR, are ideal for viral load quantification and genotyping with high sensitivity. However, they are prone to environmental contamination and false-positive results [9]. A recent method [approved by the US Federal Drug Administration (FDA) in 2014] is Roche’s COBAS HPV test for use in primary screening [10]. A major drawback of the above HPV-based tests is the high cost and the requirements for specialized experimental facilities.

Some protein biomarkers have been proposed for early cervical cancer screening. One of them is p16INK4a (cyclin-dependent kinase inhibitor) which is highly expressed at dysplastic epithelium. A combinatorial stain of p16INK4a and the cell proliferation marker Ki-67 has been proposed for increased diagnostic sensitivity [11]. However, lack of a scoring system for immunohistochemistry has hampered the incorporation of the these biomarkers in wide cervical cancer screening. Also, squamous cell carcinoma antigen (SCCA) is a known cancer antigen isolated from tissue, which is usually measured by immunoassays such as ELISA in serum or plasma patients, with limited specificity and sensitivity [12, 13].

Similarly, targets of the E5 HPV protein [e.g. epidermal growth factor receptor, p21 and p27 inhibitors of cyclin-dependent kinase, cyclooxygenase-2 (cox-2), vascular endothelial growth factor, and caveolin-1] have been proposed for early stage cervical cancer detection.  Moreover, putative markers that have been suggested are the ProEx C immunocytochemical assay (that targets the expression of topoisomerase II protein and the minichromosome maintenance complex II protein) as well as microRNAs which are regulated by E5, E6, and E7 HPV proteins [14].

Figure 1A summarizes the different methods that have been used for the diagnosis of cervical cancer.

Current proteomic studies
The limitations of the above methods demonstrate the need for the establishment of new reliable diagnostic tests via alternative methods [15]. A novel method that can expose the association of HPV infection and cellular transformation at the molecular level is proteomics [16]. Cervical cancer models (cell cultures or tissue samples) have been studied via several proteomic methods which are either gel-based [two-dimensional gel electrophoresis (2DE)] or gel-free (liquid chromatography (LC)] in combination with mass spectrometry (MS) [17]. Proteomics can be used in order to reveal putative biomarkers for early-stage diagnosis. The role of systems biology that includes the integration of proteomics data with other available ‘OMICS’ datasets, such as genomics and transcriptomics, appears to be vital towards the direction of personalized cervical cancer medicine [18].

In this review, we present some of the most recent and interesting proteomic studies for putative biomarkers in a variety of clinical samples. Diagnostic, prognostic and predictive biomarkers have been assessed in tissue, plasma/serum, cell biopsies/cervical swabs, residual fluid from cell biopsies, cell mucous and cervicovaginal fluid (CVF) by proteomic approaches [19]. Some of the most interesting studies from each category of clinical samples are reported below. The proteomics approach workflow is presented in Figure 1B.

A proteomic study, highlighted keratin 17 as a prognostic biomarker with characteristic gradual increase from normal toward cancer stage in tissue samples. Specifically, stage specific tissue samples [normal squamous mucosa, LSIL, HSIL, and SCC (total number of samples N=22] were analysed by laser capture microdissection in combination with multidimensional liquid chromatography and tandem MS (LC-MS/MS). Proteomics analysis demonstrated differentially expressed and statistically significant proteins and after bioinformatics analysis (gene ontology) keratin 4 and keratin 17 were chosen for validation by immunohistochemistry. The gradual increase from normal towards cancer stage of keratin 17 shown by proteomics, was in total agreement with the immunohistochemistry results. Kaplan–Meier curves of keratin 17 expression and general survival of cervical cancer patients revealed a strong correlation of high keratin 17 expression with poor survival, adding prognostic value to this protein [20]. Another proteomics study focused on the pelvic lymph node metastasis (PLNM) clinical status, which is important in terms of prognosis and treatment of cervical cancer. A two-dimensional difference gel electrophoresis (2D DIGE)/matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF)-MS approach was used to compare cervical tissues of patients with PLNM (N=8) to cervical tissues from patients without lymph node metastasis (NPLNM) (N=10). Analysis led to the identification of 41 differentially expressed and statistically significant proteins. Some of them (FABP5, HspB1, and MnSOD) were validated in the PLNM group compared to the NPLNM, by Western blot and immunohistochemistry [21].

The combination of Isobaric tag for relative and absolute quantitation (iTRAQ) and targeted mass spectrometric quantification was used in order to analyse serum from CIN, early- (CES), and late-stage (CLS) cervical cancer patients. A panel of six differentially expressed proteins (alpha-1-acid glycoprotein1, alpha-1-antitrypsin, serotransferrin, haptoglobin, alpha-2-HS-glycoprotein, and vitamin D-binding protein) was validated by MRM (multiple reaction monitoring) in an independent set of 229 serum samples consisting of controls (N=49), CIN-1 to 3 designated as CIN (N=48), CES (N = 49), CLS (N=34), and ovarian cancer (N=49). The above panel discriminated patients with CIN from healthy controls with a sensitivity of 67% and specificity of 88%. Combination of the specific panel with SCCA, a well-studied putative biomarker for cervical cancer, improved discrimination of CIN, CES, and CLS patients from healthy control. Briefly, upon the comparison of the CES versus healthy group, the area under the curve was 0.86 (sensitivity/specificity = 71/90%), when using the six-protein panel and SCCA [12].

An alternative strategy was used by Boylan et al., in order to study the proteome of Pap test clinical samples. The cell-free residual fluid from cell biopsies was collected, proteins were isolated (acetone precipitation) and their concentration upon resolubilization was determined by Bradford assay. Filter aided sample preparation followed by LC-MS/MS analysis yielded 300 protein identifications per sample and 700 unique protein identifications after pooling the samples. Many of the proteins had similar biological functions to the ones identified from CVF. Thus, residual fluid could be an alternative material for the study of proteins related to cervical dysplasia [22].

The cell mucous proteome from 25 HPV-positive and pre invasive cervical disease samples has been investigated by a combination of 2DE MS, gel-based LC-MS/MS and 2DE MS after depletion of highly abundant proteins (e.g. albumin). The above approaches were combined and 107 unique proteins were identified. A bioinformatics study showed that they are related to metabolism, immune response, and transport. Proteins like acute-phase plasma proteins, α-1-antichymotrypsin and α-1-antitrypsin, were found to be both phosphorylated and glycosylated after posttranslational modifications evaluation with appropriate fluorescent dyes [23].

Table 1 highlights the above clinical proteomics studies and some of the most promising putative biomarkers that were identified.

Discussion and conclusions
Cervical cancer is the fourth most common cancer in women. The Pap test is a very efficient diagnostic approach in terms of specificity (77.8-98.8%) but has a variable sensitivity (33.8% to 94.0%) [8). Molecular tests such as HPV DNA detection by PCR and/or hybridization with adequate probes are the reference methods for the detection of HPV. The aforementioned methods are often expensive and unavailable in the developing regions where cervical cancer is very frequent (85% of cervical cancer cases). A promising idea is that proteomics will facilitate the discovery of novel biomarkers that will enable the future cervical cancer screening in developing countries in the form of antibody-based practical diagnostic self-tests (like pregnancy tests). New and advanced proteomic techniques like MRM could validate several biomarkers that will eventually be combined into panels for more accurate testing in the above diagnostic tests. Of course, the future of proteomics studies is not only promising but also challenging. New aspects of research should be taken into consideration.  Redox proteomics will be used for the exploration of proteins oxidation status in order to reveal the interaction of oxidative stress and tumour development. In particular, the oxidative status of proteins in HPV-related cervical cancer cells was explored via oxidative isotope-coded affinity tags (OxICAT) and voltage-dependent anion channel 1 (VDAC1) was found to be highly oxidized in HPV-positive cervical cancer cells [24]. The important role of post-translational modifications (PTMs) such as phosphorylation and glycosylation should be thoroughly examined. The combination of systems biology and proteomics offers the possibility to elucidate cervical cancer mechanisms and identify potential biomarkers for early-stage detection.

References
1. Pappa KI, Polyzos A, Jacob-Hirsch J, Amariglio N, Vlachos GD, Loutradis D, Anagnou NP. Profiling of discrete gynecological cancers reveals novel transcriptional modules and common features shared by other cancer types and embryonic stem cells. PloS One 2015; 10(11): e0142229.
2. Doorbar J, Quint W, Banks L, Bravo IG, Stoler M, Broker TR, Stanley MA. The biology and life-cycle of human papillomaviruses. Vaccine 2012; 30(Suppl 5): F55–70.
3. Schiffman MH, Castle P. Epidemiologic studies of a necessary causal risk factor: human papillomavirus infection and cervical neoplasia. J Natl Cancer Inst. 2003; 95(6): E2.
4. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015; 136(5): E359–386.
5. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011; 61(2): 69–90.
6. Wilting SM, Smeets SJ, Snijders PJ, van Wieringen WN, van de Wiel MA, Meijer GA, Ylstra B, Leemans CR, Meijer CJ, et al. Genomic profiling identifies common HPV-associated chromosomal alterations in squamous cell carcinomas of cervix and head and neck. BMC Med Genomics 2009; 2: 32.
7. Nayar R, Wilbur DC. The Pap test and Bethesda 2014. Cancer Cytopathol. 2015; 123(5): 271–281.
8. Wright TC, Jr. HPV DNA testing for cervical cancer screening. FIGO 26th Annual Report on the Results of Treatment in Gynecological Cancer. Int J Gynaecol Obstet. 2006; 95(Suppl 1): S239–246.
9. Hubbard RA. Human papillomavirus testing methods. Arch Pathol Lab Med. 2003; 127(8): 940–945.
10. Hogarth S, Hopkins M, Rotolo D. Technological accretion in diagnostics: HPV testing and cytology in cervical cancer screening. In: Consoli D, Mina A, Nelson RR, Ramlogan R (eds) Medical Innovation: Science, Technology and Practice. Routledge 2015.
11. von Knebel Doeberitz M, Reuschenbach M, Schmidt D, Bergeron C. Biomarkers for cervical cancer screening: the role of p16(INK4a) to highlight transforming HPV infections. Expert Rev Proteomics 2012; 9(2): 149–163.
12. Boichenko AP, Govorukhina N, Klip HG, van der Zee AG, Guzel C, Luider TM, Bischoff R. A panel of regulated proteins in serum from patients with cervical intraepithelial neoplasia and cervical cancer. J Proteome Res. 2014; 13(11): 4995–5007.
13. Kato H, Torigoe T. Radioimmunoassay for tumor antigen of human cervical squamous cell carcinoma. Cancer 1977; 40(4): 1621–1628.
14. de Freitas AC, Coimbra EC, Leitao Mda C. Molecular targets of HPV oncoproteins: potential biomarkers for cervical carcinogenesis. Biochim Biophys Acta 2014; 1845(2): 91–103.
15. Wentzensen N, von Knebel Doeberitz M. Biomarkers in cervical cancer screening. Dis Markers 2007; 23(4): 315–330.
16. Lomnytska M, Souchelnytskyi S. Markers of breast and gynecological malignancies: the clinical approach of proteomics-based studies. Proteomics Clin appl. 2007; 1(9): 1090–1101.
17. Di Domenico F, De Marco F, Perluigi M. Proteomics strategies to analyze HPV-transformed cells: relevance to cervical cancer. Expert Rev Proteomics 2013; 10(5): 461–472.
18. Breuer EK, Murph MM. The role of proteomics in the diagnosis and treatment of women’s cancers: current trends in technology and future opportunities. Int J Proteomics 2011; 2011: pii: 373584.
19. Kontostathi G, Zoidakis J, Anagnou NP, Pappa KI, Vlahou A, Makridakis M. Proteomics approaches in cervical cancer: focus on the discovery of biomarkers for diagnosis and drug treatment monitoring. Exp Rev Proteomics 2016; 13(8): 731–745.
20. Escobar-Hoyos LF, Yang J, Zhu J, Cavallo JA, Zhai H, Burke S, Koller A, Chen EI, Shroyer KR. Keratin 17 in premalignant and malignant squamous lesions of the cervix: proteomic discovery and immunohistochemical validation as a diagnostic and prognostic biomarker. Mod Pathol. 2014; 27(4): 621–630.
21. Wang W, Jia HL, Huang JM, Liang YC, Tan H, Geng HZ, Guo LY, Yao SZ. Identification of biomarkers for lymph node metastasis in early-stage cervical cancer by tissue-based proteomics. Br J Cancer 2014; 110(7): 1748–1758.
22. Boylan KL, Afiuni-Zadeh S, Geller MA, Hickey K, Griffin TJ, Pambuccian SE, Skubitz AP. A feasibility study to identify proteins in the residual Pap test fluid of women with normal cytology by mass spectrometry-based proteomics. Clin Proteomics 2014; 11(1): 30.
23. Panicker G, Ye Y, Wang D, Unger ER. Characterization of the human cervical mucous proteome. Clini Proteomics 2010; 6(1–2): 18–28.
24. Zhang C, Ding W, Liu Y, Hu Z, Zhu D, Wang X, Yu L, Wang L, Shen H, et al. Proteomics-based identification of VDAC1 as a tumor promoter in cervical carcinoma. Oncotarget 2016; doi: 10.18632/oncotarget.10562 [Epub ahead of print].

The authors
Georgia Kontostathi1,2 MSc; Jerome Zoidakis1 PhD; Nicholas P. Anagnou2,3 MD, PhD; Kalliopi I. Pappa3,4 MD, PhD; Manousos Makridakis*1 PhD
1Biotechnology Division, Biomedical Research Foundation, Academy of Athens (BRFAA), Athens, Greece
2Laboratory of Biology, University of
Athens School of Medicine, Athens, Greece
3Cell and Gene Therapy Laboratory,
Biomedical Research Foundation,
Academy of Athens (BRFAA), Athens, Greece
4First Department of Obstetrics and
Gynecology, University of Athens School of Medicine, Athens, Greece

*Corresponding author
E-mail: mmakrid@bioacademy.gr

C287 Tang CLI Figure 1

RAS pathway biomarkers for breast cancer prognosis

Metastatic breast cancer is a highly heterogeneous, rapidly evolving and devastating disease that challenges our ability to find curative therapies. RAS pathway activation is an understudied research area in breast cancer. EGFR/RAS pathway activation is prevalent in breast cancer with poor prognosis. The prognostic RAS pathway biomarkers can be used to identify resistant tumour clones, stratify patients and guide therapies.

by L. L. Siewertsz van Reesema, M. P. Lee, Dr V. Zheleva, Dr J. S. Winston,
Dr C. F. O’Connor, Prof. R. R. Perry, Prof. R. A. Hoefer and Prof. A. H. Tang

Introduction
Breast cancer currently represents the second leading cause of cancer-related deaths among women in the United States [1]. In 2016 alone, an estimated 246 660 women will be diagnosed and an additional 40 450 women are expected to succumb to the disease process [1, 2]. By 2030, there are projected to be 294 000 new cases, making breast cancer a growing public health concern [3–5]. The increased breast cancer screenings, major technical advancements in breast cancer imaging and early detection, and targeted anti-cancer therapies have contributed to significant decreases in morbidity and mortality since the 1970s, and now more than 90% of patients with early-stage breast cancer have expected survival of much more than 5 years [2, 5]. Despite these amazing advancements, the prognosis for patients with high-grade, locally advanced and metastatic breast cancers remains poor with average survival less than 2 years [2, 6–8]. State-of-the-art treatments, such as anti-HER2 therapy, anti-ER therapy, anti-PI3K and anti-mTOR therapy, tumour genome-guided combination therapies, stem cell therapy, and anti-CTLA-4/PD1 immunotherapy, alone or in combination, are not curative in eradicating metastatic breast cancer [9–14]. The clinical reality is that despite similar clinical diagnoses and presentation, patients often display diverse tumour response to standard therapies. The intrinsic diversity and evolving heterogeneity of their mammary tumours can become more pronounced in relapsed or metastatic tumours after therapeutic intervention and clonal selection by ineffective treatment. This de novo and acquired tumour heterogeneity leads to diverse tumour responses in neoadjuvant and adjuvant setting following standard-of-care therapies, which in turn leads to varied clinical outcome and disparity in patient survival [6, 15, 16].

Currently, there are no reliable clinical biomarkers that can be used to consistently predict survival of patients with late-stage or metastatic breast cancers [6]. Classically utilized breast cancer biomarkers, such as estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) molecular subtypes, do not correlate with survival outcomes nor do they predict tumour responses to guided therapies in invasive disease [6]. Patients with malignant tumours are often subjected to full regimens of toxic therapies followed by a period of uncertainty awaiting to learn the response of their tumours and efficacy of their treatments. Unfortunately, some patients may be over-treated, which compromises their long-term quality of life, or, conversely, some patients may be under- or incorrectly treated and thus miss a critical window of opportunity to change prescribed anticancer regimens if therapy-resistant tumours persist after aggressive therapy. There is a pressing need to identify reliable, sensitive, and robust prognostic biomarkers to closely monitor tumour response in real-time, and, importantly, to determine whether first-line standard therapies prescribed are effective against high-risk and invasive mammary tumours, given diverse tumour response and intrinsic tumour heterogeneity [7, 16–21].

Current prognostic biomarkers used in breast cancer
Clinicopathological parameters such as patient age, TMN (tumour size, lymph node status, metastasis) staging, tumour grade and histology, and molecular subtype of breast tumours have become commonplace in justifying medical decision making and prescribing treatment modalities. Therapies for invasive and high-risk breast cancer (luminal, basal-like, HER2-positive and triple-negative breast cancer) are routinely selected based on tumour ER, PR and HER2 status, in addition to aforementioned clinicopathological parameters [22–25]. The American Society of Clinical Oncology (ASCO) updated their clinical practice guidelines this year and according to the panel, only the current gold-standard biomarkers ER, PR, and HER2 should be used to guide treatment decisions, despite the fact they do not correlate with survival outcomes, nor predict tumour response to standard-of-care therapies [26].

ER-positive breast tumours account for the majority of cases worldwide [27]. Estrogen is the main stimulation for growth in the mammary tumours allowing this clinical biomarker to direct treatment planning and assess the benefit of mainstay anti-hormone therapies such as tamoxifen and aromatase inhibitors. The expression of PR is frequently associated with ER-positive tumours where less than 1% of mammary tumours are PR-positive and ER negative [27]. Furthermore, PR expression has shown strong prognostic value with high level PR expression in invasive breast tumours displaying better outcomes to anti-hormone treatment when compared to low level PR expression [28]. Current guidelines put forth by the ASCO recommend ER and PR testing of all new cases of primary and distant recurrent breast tumours [29].

HER2 status has been identified as a strong indicator of patient overall survival. Overexpression of the HER2 oncogene leads to higher chances of relapse with shorter survival times. The development of anti-HER2 therapies, such as trastuzumab, in treating HER2 have been shown to have major benefit and therefore HER2 status is tested in the majority of breast cancer diagnoses [30]. Other emerging biomarkers such as Ki67 have a possible prognostic role in invasive breast cancers [31]. While the exact function is unknown, Ki67 is expressed in cycling tumour cells and it has strong correlation with tumour grade [31, 32]. Approaches to combining Ki67 with established biomarkers ER, PR, and HER2 are now being used to further distinguish breast cancers into clinical subtypes. However, the ASCO did not recommend using Ki67 to guide adjuvant therapies [29].

The ASCO has recognized the clinical utility of six multigene expression assays for specific subgroups of patients, including OncotypeDX (Genomic Health), EndoPredict (Sividon Diagnostics), PAM50 (Prosigna Breast Cancer Prognostic Gene Signature Assay), Breast Cancer Index, urokinase plasminogen activator, and plasminogen activator inhibitor type 1 [33–37]. Most of these assays are recommended for patients with early-stage breast cancers only; none of these multi-gene assays were encouraged for patients with HER2-positive or triple-negative cancers, in addition to late-stage or metastatic cancers [34, 35]. The development of these promising gene signature-based molecular assessment tools for breast cancer is encouraging for the personalization of breast cancer therapies; however, there still remains an unmet medical need for patients with locally advanced and metastatic breast cancers.

The potential role of RAS pathway proteins in the prognosis and treatment of breast cancer
The oncogenic K-RAS carries the most common gain-of-function mutations in human cancer (30% of all human cancers) and its oncogenic role has been well-established in many types of human cancers [38, 39]. Thus, focusing on this tumour-promoting RAS signalling pathway is a logical choice of investigation for prognostic biomarker discovery and novel anticancer therapy development against oncogenic K-RAS-driven human cancer [40, 41]. The RAS pathway has been an intensely studied area of cancer research due to its activation of downstream effectors resulting in tumour proliferation, survival and motility (Fig. 1A). The loss of its most essential downstream signalling gatekeeper, SIAH (seven in absentia homologue), impeded oncogenic K-RAS-driven human pancreatic cancer and lung cancer [40, 42, 43]. Although oncogenic K-RAS mutations are rarely detected in mammary tumours (observed in about 5% of patients), genomic studies have indicated that the EGFR/HER2/K-RAS ‘pathway’ is activated in a large proportion of aggressive and malignant breast cancers [44, 45]. EGFR/HER2/K-RAS activation has been correlated with shortened survival, resistance to therapy, and tumour relapse despite aggressive treatments in breast cancer [44–47]. The mechanism and function of persistent RAS pathway activation remains elusive in breast cancer. This lack of mechanistic understanding, along with the low mutation rate of oncogenic RAS/RAF/MARK detected in mammary tumours, may contribute to the tumour-driving RAS pathway activation being understudied in the area of breast cancer research. It should also be noted that activation of RAS pathway in mammary tissues of animal models is adequate to induce oncogenic transformation and malignancy [48, 49]. Therefore, although there may not be any detectable RAS mutations in high-grade, locally advanced and metastatic breast tumours, alternative RAS pathway activation mechanisms of the RAS pathway downstream effectors may be present, promoting mammary tumorigenesis and metastasis.

Since RAS pathway activation is a major and essential signalling hub to promote human malignancies, we investigated whether EGFR/HER2/RAS pathway biomarker expression can be added to evaluate therapy efficacy and predict patient survival in breast cancer (Fig. 1A). It has been established that expression of SIAH, the most downstream signalling module and the most evolutionarily conserved component of the RAS signalling pathway, reflects RAS pathway activation, cell proliferation, and tumour growth [40, 42, 43]. In a retrospective study, 364 matched pairs of breast tumour specimens from 182 patients who received neoadjuvant systemic therapy (NST) were analysed to determine whether SIAH and/or EGFR outperform ER, PR, HER2, and Ki67 as a prognostic biomarker in locally advanced and metastatic breast cancer. The prognostic power of SIAH and EGFR, alone or in combination, is comparable to the clinical gold standards of clinical predictors (lymph node positivity, mammary tumour size, grade, stage and molecular subtypes in combination), and imaging-guided technology [50]. The activation/inactivation of the tumour-promoting RAS pathway biomarkers, SIAH and EGFR, is associated with tumour progression/regression in mammary tumours post-neoadjuvant systemic therapies (NST). We found that a marked reduction in SIAH/EGFR expression post-NST would indicate effective therapy and increased survival, while persistent high SIAH/EGFR expression post-NST would indicate ineffective therapy and decreased survival [50]. These results suggest that NST-induced reduction of SIAH and EGFR expression may be used as two useful surrogate prognostic biomarkers to quantify therapeutic efficacy, determine tumour responses, detect emerging resistant clones, and predict patient survival in high-grade and aggressive molecular subtype of breast cancer in the neoadjuvant setting in the future (Fig. 1B).

Conclusion
Early stage breast cancer is highly responsive to commonly prescribed standard of care therapies with excellent long-term survival. Locally advanced and metastatic breast cancer has a much worse prognosis despite aggressive chemo- and radiation therapies and locoregional surgical interventions. This disparity in prognosis underlines the acute need to tailor therapy and stratify patients in order to improve patient survival. The discovery of incorporating tumour heterogeneity-independent, therapy-responsive and tumour-driven RAS pathway biomarkers is prognostic in breast cancer, have a clear clinical impact to benefit breast cancer patients with locally advanced and metastatic diseases.

References
1. Siegel RL, et al. Cancer statistics, 2016. CA Cancer J Clin. 2016; 66: 7–30.
2. DeSantis C, et al. Breast cancer statistics, 2013. CA Cancer J Clin. 2014; 64: 52–62.
3. Anderson WF,  et al. Incidence of breast cancer in the United States: current and future trends. J Natl Cancer Inst. 2011; 103: 1397–1402.
4. Rahib L, et al. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014; 74: 2913–2921.
5. Graham LJ, et al. Current approaches and challenges in monitoring treatment responses in breast cancer. J Cancer 2014; 5: 58–68.
6. Tevaarwerk AJ, et al. Survival in patients with metastatic recurrent breast cancer after adjuvant chemotherapy: little evidence of improvement over the past 30 years. Cancer 2013; 119: 1140–1148.
7. Zardavas D, et al. Emerging targeted agents in metastatic breast cancer. Nat Rev Clin Oncol. 2013; 10: 191–210.
8. Lobbezoo DJ, et al. Prognosis of metastatic breast cancer subtypes: the hormone receptor/HER2-positive subtype is associated with the most favorable outcome. Breast Cancer Res Treat. 2013; 141: 507–514.
9. El Saghir NS, et al. Treatment of metastatic breast cancer: state-of-the-art, subtypes and perspectives. Crit Rev Oncol Hematol. 2011; 80: 433–449.
10. Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 2009; 9: 550–562.
11. McKeage K, Perry CM. Trastuzumab: a review of its use in the treatment of metastatic breast cancer overexpressing HER2. Drugs 2002; 62: 209–243.
12. Piccart-Gebhart MJ, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005; 353: 1659–1672.
13. Robert N, et al. Randomized phase III study of trastuzumab, paclitaxel, and carboplatin compared with trastuzumab and paclitaxel in women with HER-2-overexpressing metastatic breast cancer. J Clin Oncol. 2006; 24: 2786–2792.
14. Romond EH, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005; 353: 1673–1684.
15. Vogelstein B, et al. Cancer genome landscapes. Science 2013; 339: 1546–1558.
16. Zardavas D, et al. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol. 2015; 12: 381–394.
17. Coley HM. Mechanisms and strategies to overcome chemotherapy resistance in metastatic breast cancer. Cancer Treat Rev. 2008; 34: 378–390.
18. Haddad TC, Goetz MP. Landscape of neoadjuvant therapy for breast cancer. Ann Surg Oncol. 2015; 22: 1408–1415.
19. Holohan C, et al. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 2013; 13: 714–726.
20. Hutchinson L. Breast cancer: challenges, controversies, breakthroughs. Nat Rev Clin Oncol. 2010; 7: 669–670.
21. King TA, Morrow M. Surgical issues in patients with breast cancer receiving neoadjuvant chemotherapy. Nat Rev Clin Oncol. 2015; 12: 335–343.
22. Baselga J, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012; 366: 109–119.
23. Bevers TB, et al. NCCN clinical practice guidelines in oncology: breast cancer screening and diagnosis. J Natl Compr Canc Netw. 2009; 7: 1060–1096.
24. Redden MH, Fuhrman GM. Neoadjuvant chemotherapy in the treatment of breast cancer. Surg Clin North Am. 2013; 93: 493–499.
25. Tolaney SM, et al. Adjuvant paclitaxel and trastuzumab for node-negative, HER2-positive breast cancer. N Engl J Med. 2015; 372: 134–141.
26. Hammond ME, et al. American Society of Clinical Oncology/College Of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol. 2010; 28: 2784–2795.
27. Viale G, et al. Prognostic and predictive value of centrally reviewed expression of estrogen and progesterone receptors in a randomized trial comparing letrozole and tamoxifen adjuvant therapy for postmenopausal early breast cancer: BIG 1–98. J Clin Oncol. 2007; 25: 3846–3852.
28. Dowsett M, et al. Benefit from adjuvant tamoxifen therapy in primary breast cancer patients according oestrogen receptor, progesterone receptor, EGF receptor and HER2 status. Ann Oncol. 2006; 17: 818–826.
29. Runowicz CD, et al. American Cancer Society/American Society of Clinical Oncology Breast Cancer Survivorship Care Guideline. J Clin Oncol. 2016; 34: 611–635.
30. Weigel MT, Dowsett M. Current and emerging biomarkers in breast cancer: prognosis and prediction. Endocr Relat Cancer 2010; 17: R245–262.
31. Falato C, et al. Ki67 measured in metastatic tissue and prognosis in patients with advanced breast cancer. Breast Cancer Res Treat. 2014; 147: 407–414.
32. Bulfoni M, et al. In patients with metastatic breast cancer the identification of circulating tumor cells in epithelial-to-mesenchymal transition is associated with a poor prognosis. Breast Cancer Res. 2016; 18: 30.
33. Sparano JA, et al. Prospective validation of a 21-gene expression assay in breast cancer. N Engl J Med. 2015;.
34. Gyorffy B, et al. Multigene prognostic tests in breast cancer: past, present, future. Breast Cancer Res. 2015; 17: 11.
35. Goncalves R, Bose R. Using multigene tests to select treatment for early-stage breast cancer. J Natl Compr Canc Netw. 2013; 11: 174–182; quiz 182.
36. van ‘t Veer LJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002; 415: 530–536.
37. Oakman C, et al. Breast cancer assessment tools and optimizing adjuvant therapy. Nat Rev Clin Oncol. 2010; 7: 725–732.
38. Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003; 3: 11–22.
39. Pylayeva-Gupta Y, et al. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer 2011; 11: 761–774.
40. Van Sciver RE, et al. Blocking SIAH proteolysis, an important K-RAS vulnerability, to control and eradicate K-RAS-driven metastatic cancer. In: A. Azmi (Ed.) Conquering RAS, Elsevier, 2016.
41. McCormick F. KRAS as a therapeutic target. Clin Cancer Res. 2015; 21: 1797–1801.
42. Ahmed AU, et al. Effect of disrupting seven-in-absentia homolog 2 function on lung cancer cell growth. J Natl Cancer Inst. 2008; 100: 1606–1629.
43. Schmidt RL, et al. Inhibition of RAS-mediated transformation and tumorigenesis by targeting the downstream E3 ubiquitin ligase seven in absentia homologue. Cancer Res. 2007; 67: 11798–11810.
44. Arteaga CL, et al. Treatment of HER2-positive breast cancer: current status and future perspectives. Nat Rev Clin Oncol. 2012; 9: 16–32.
45. Foulkes WD, et al. Triple-negative breast cancer. N Engl J Med. 2010; 363: 1938–1948.
46. Tebbutt N, et al. Targeting the ERBB family in cancer: couples therapy. Nat Rev Cancer 2013; 13: 663–673.
47. Wright KL, et al. Ras signaling is a key determinant for metastatic dissemination and poor survival of luminal breast cancer patients. Cancer Res. 2015; 75: 4960–4972.
48. Creedon H, et al. Use of a genetically engineered mouse model as a preclinical tool for HER2 breast cancer. Dis Model Mech. 2016; 9: 131–140.
49. Politi K, Pao W. How genetically engineered mouse tumor models provide insights into human cancers. J Clin Oncol. 2011; 29: 2273–2281.
50. van Reesema LL, et al. SIAH and EGFR, two RAS pathway biomarkers, are highly prognostic in locally advanced and metastatic breast cancer. EBioMedicine 2016; 11: 183–198.

The authors
Lauren L. Siewertsz van Reesema†*1 BS; Michael P. Lee†1 MS; Vasilena Zheleva2 MD; Janet S. Winston3 MD; Caroline F. O’Connor4 MD; Roger R. Perry2 MD, FACS; Richard A. Hoefer5,6,7 DO, FACS; Amy H. Tang*4,8 PhD
1Eastern Virginia Medical School, Norfolk, VA 23507, USA
2Department of Surgery, Eastern Virginia Medical School, Norfolk, VA 23507, USA
3Sentara Pathology and Pathology Sciences Medical Group, Sentara Norfolk General Hospital (SNGH), Norfolk, VA 23507, USA
4Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
5Sentara Cancer Network, 6Dorothy G. Hoefer Comprehensive Breast Center, 7Sentara CarePlex Hospital, Newport News, Virginia 23606, USA
8Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA

†Authors contributed equally to this publication.
*Corresponding author
E-mail: Tangah@evms.edu

C285 Figure 1

New developments in the early diagnosis of ovarian cancer

Ovarian cancer is difficult to diagnose early, with consequent poor survival. Evidence suggests many cases may originate in precursor lesions in the fallopian tubes. Differential expression of specific proteins in the fallopian tubes of women with high-grade serous ovarian cancer, detected by immunohistochemistry, shows promise as a potential novel diagnostic marker.

by Dr Kezia Gaitskell and Prof. Ahmed Ashour Ahmed

Background
Ovarian cancer is the seventh greatest cause of cancer mortality amongst women worldwide, and the fifth greatest cause amongst women in more developed regions [1]. In the USA, 60% of women with ovarian cancer already have distant metastases at diagnosis, for which the 5-year survival is less than 30% [2]. Early clinical diagnosis of ovarian cancer is difficult, as symptoms are often non-specific, such as abdominal distention, urinary frequency, or abdominal pain [3].

Current evidence on ovarian cancer diagnosis and screening
Diagnostic investigations include imaging (e.g. ultrasound, CT or MRI of the pelvis and abdomen), together with blood tests for tumour markers – particularly cancer antigen 125 (CA-125) [4]. However, although CA-125 is the main biomarker used in the diagnosis of ovarian cancer, it is far from perfect in sensitivity and specificity: although approximately 80% of women with epithelial ovarian cancer will have a CA-125 concentration above the cut-off value of 35 IU/mL, CA-125 may also be elevated with other cancers (including liver, pancreatic, lung, and endometrial cancers), and physiological or benign conditions (including menses, pregnancy, cirrhosis, salpingitis, pancreatitis and endometriosis) [5].

A variety of additional putative tumour markers have been suggested for use in combination with CA-125, but current guidelines from the National Comprehensive Cancer Network in the USA are that there is insufficient evidence for their usefulness in detecting early-stage ovarian cancer [4], although the European Group on Tumor Markers suggests that human epididymis protein 4 (HE4) may be helpful for the differential diagnosis of pelvic masses, particularly in premenopausal women [5].

There has been considerable interest in finding markers of early disease, that could enable earlier diagnosis or screening for ovarian cancer. Two large randomized controlled trials have been performed of screening for ovarian cancer: the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (PLCO) in the USA [6], and the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) in the UK [7]. Both trials randomized women to either no screening, or screening with the CA-125 blood test with or without trans-vaginal ultrasound. Unfortunately, neither trial could demonstrate a clear mortality benefit with screening, although there was a suggestion of benefit in some secondary analyses in the UKCTOCS trial [7].

New hypotheses of the origins of ovarian cancer
The search for potential early markers of ovarian cancer is also affected by the increasing evidence of heterogeneity between the tumour subtypes. Ovarian cancer has traditionally been divided into subtypes on the basis of microscopic morphology, the most common types being serous, endometrioid, clear cell, and mucinous tumours. There is growing evidence that these different histological tumour subtypes have different characteristic genetic mutations, and may have distinct origins [8, 9]. In particular, there is evidence that many cases of high-grade serous ovarian cancer (the most common subtype) may arise from precursor lesions in the fallopian tube epithelium, known as serous tubal intraepithelial carcinoma (STIC) (reviewed by Nik et al. [10]). These STIC lesions show dysplastic morphological changes, and also tend to show the mutations in the tumour-suppressor gene TP53 that are characteristic of high-grade serous ovarian carcinoma, and increased expression of the proliferation marker Ki-67. There is also evidence that some cases of endometrioid and clear cell ovarian cancer (less common subtypes) may arise from endometriosis (reviewed by Munksgaard & Blaakaer [11]). The origins of low-grade serous tumours and mucinous carcinomas are less certain, although several hypotheses exist.

The hypothesis that many, if not most, high-grade serous ‘ovarian’ cancers may in fact arise from the fallopian tubes has led to increasing interest in exploring changes in the fallopian tubes as potential early markers. The discovery of STIC lesions is interesting in terms of improving our understanding of pathogenesis, but is not currently useful for identifying changes early in malignancy, or pre-malignancy, in clinical practice. One limitation is that STIC lesions tend to be very focal, and are most common at the fimbrial end of the fallopian tubes, adjacent to the ovary, which is difficult to access without surgical removal of the fallopian tubes.

New findings regarding the role of SOX2
We investigated increased expression of SOX2, a key stem cell differentiation gene, as a possible marker of high-grade serous carcinogenesis within the fallopian tubes. We chose SOX2 because work from our group had shown that mutations at several sites near the SOX2 gene were ubiquitous in samples of ovarian cancer taken from multiple locations and time points in a single patient, indicating that they acted as early ‘driver’ mutations [12]. We showed that SOX2 expression (detected using immunohistochemistry) was significantly increased in the fallopian tube epithelial cells of women with high-grade serous ovarian cancer, compared to women with endometrial cancer or benign disease (e.g. uterine fibroids) [12], as illustrated in Figure 1.

We also found that SOX2 expression in the fallopian tubes was significantly increased in women with germline mutations in the tumour suppressor genes BRCA1 and BRCA2, who are known to be at higher risk of breast and ovarian cancer [12]. These women with BRCA1/2 mutations had their ovaries and fallopian tubes removed to reduce their subsequent risk of cancer, but did not have evident cancer at the time of surgery. Thus, the finding that elevated SOX2 expression was detectable in their fallopian tubes suggests that increased SOX2 expression may be an early sign of precancerous changes within the fallopian tubes.

Potential future implications
Our observation that SOX2 expression is increased in the fallopian tube epithelial cells of women with high-grade serous ovarian cancer, and women with BRCA1/2 mutations, compared to women with other cancers or benign disease, suggests that SOX2 might have a potential role as a biomarker in the early diagnosis of ovarian cancer. However, several challenges remain before testing for SOX2 expression could be considered in clinical practice – particularly the anatomical difficulty of sampling the fallopian tube epithelium without invasive surgery, and the fact that SOX2 is a nuclear marker. Our research group is currently exploring other potential cell-surface markers that correlate with SOX2 expression, which might be easier to detect.

Summary
There are many challenges in the early diagnosis of ovarian cancer. New evidence of the possible tubal origins of high-grade serous ovarian cancer is changing the approach to identifying potential new biomarkers of early disease. SOX2 has emerged as a promising marker, but further work is needed before it would be suitable for routine clinical practice.

References
1. Ferlay J, Soerjomataram I, et al. GLOBOCAN 2012 v1.0: Estimated cancer incidence and mortality worldwide in 2012. International Agency for Research on Cancer/World Health Organization 2013 (http://globocan.iarc.fr/Default.aspx).
2. Howlader N, Noone AM, et al. Cronin KA (eds). SEER Cancer Statistics Review, 1975-2013. National Cancer Institute, Bethesda, MD, USA 2016 (http://seer.cancer.gov/csr/1975_2013/).
3. Hamilton W, Peters TJ, et al. Risk of ovarian cancer in women with symptoms in primary care: population based case-control study. BMJ 2009; 339: b2998.
4. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines): Ovarian cancer including fallopian tube cancer and primary peritoneal cancer, Version 1.2016. Ft. Washington, PA, USA. National Comprehensive Cancer Network 2016 (https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf).
5. Soletormos G, Duffy MJ, et al. Clinical use of cancer biomarkers in epithelial ovarian cancer: updated guidelines from the European group on tumor markers. Int J Gynecol Cancer 2016; 26(1): 43–51.
6. Buys SS, Partridge E, et al. Effect of screening on ovarian cancer mortality: the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Randomized Controlled Trial. JAMA 2011; 305(22): 2295–2303.
7. Jacobs IJ, Menon U, et al. Ovarian cancer screening and mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): a randomised controlled trial. Lancet 2016; 387(10022): 945–956.
8. Kurman RJ, Shih IeM. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer–shifting the paradigm. Hum Pathol. 2011; 42(7): 918–931.
9. Prat J. Ovarian carcinomas: five distinct diseases with different origins, genetic alterations, and clinicopathological features. Virchows Arch. 2012; 460(3): 237–249.
10. Nik NN, Vang R, et al. Origin and pathogenesis of pelvic (ovarian, tubal, and primary peritoneal) serous carcinoma. Annu Rev Pathol. 2014; 9: 27–45.
11. Munksgaard PS, Blaakaer J. The association between endometriosis and ovarian cancer: a review of histological, genetic and molecular alterations. Gynecol Oncol. 2012; 124(1): 164–169.
12. Hellner K, Miranda F, et al. Premalignant SOX2 overexpression in the fallopian tubes of ovarian cancer patients: discovery and validation studies. EBioMedicine 2016; 10: 137–149.

The authors
Kezia Gaitskell*1,2 BM BCh; Ahmed Ashour Ahmed1,2 MBBCh, MRCOG, PhD
1Ovarian Cancer Cell Laboratory, Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford OX3 9DS, UK
2Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Women’s Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK

*Corresponding author
E-mail: Kezia.gaitskell@ceu.ox.ac.uk

C268 mass spectrometry tosh thematic crop

Mass spectrometry – small samples, high-speed, low cost

Recent years have witnessed the growing use of mass spectrometry (MS) in the clinical laboratory. MS provides massive improvements in the sensitivity and specificity of clinical tests. It does this by using an ionized molecule’s mass/charge (m/z) ratio for identification.
MS has its roots in the screening and fingerprinting of molecules in drugs of abuse. Over the years, the technology has rapidly evolved. Today, it is routinely used to screen for diseases and to precisely identify causes of infections for targeted therapies. The analysis of proteins is also accelerating, with special potential demonstrated by biomarkers such as thyroglobulin.Alongside, limitations to immunoassays have also driven adoption of MS. For example, no immunoassays were approved for the immunosuppressant sirolimus and this compelled laboratories to turn to MS. Another advantage lay in improved assay quality, such as the measurement of testosterone in patients with low endogenous concentrations, such as women and children.

Enticing advantages
One of the most enticing set of advantages of mass spectrometry is that it provides clinically relevant information from relatively small sample volumes, and does this both rapidly and at a reduced cost. Gas chromatography (GC), liquid chromatography (LC) and ion mobility spectrometry (IMS) separation now allow targeting of ever-smaller analyte concentrations. LC-MS/MS (liquid chromatography-tandem mass spectrometry), on its part, offers scope to cut costs further, while continuing to improve accuracy. Other technology trends include integrating MS with low-flow chromatography, ultra-high pressure chromatography and online/multi-dimensional  chromatography.
Nevertheless, MS also adds a new layer of complexity. As a result, close and well-structured communication between laboratories and clinicians is a vital component for the effective use of MS.

A three-step process
Today, there are three principal steps for conducting an analysis by MS: sample preparation, separation by gas-chromatography (GC) or liquid-chromatography (LC), and mass spectrometric analysis.
In MS, a sample ‘matrix’ refers to everything present in a sample, excluding analytes of interest. Differences in behaviour between analytes and matrix components determines the choice of sample preparation. Although sample preparation requires more labour than immunoassays, in-house mass spectrometry-based assays are now considered cost-effective, even for smaller labs.

Sample preparation
The preparation of samples and their subsequent separation by chromatography both use mechanisms which first position molecules (the stationary phase) and then separate analytes from matrix components (the mobile phase).
Preparation firstly depends on the sample type selected for analysis (e.g. blood/serum or urine). Analytes from serum (including blood fractions) require the maximum care in preparation, owing to a relatively low ratio in the concentration of analytes to matrix components. On the other hand, urine analytes are often compatible with simple dilution protocols, due to the concentrating effect of kidneys in the production of urine.

Typical techniques in preparing samples include solid-phase extraction (SPE), immunoextraction and dilution. The choice depends principally on whether the analytes are acidic or alkaline, and if they are heavily protein-bound.

Solid-phase extraction
SPE is based on combining a solid stationary phase with a liquid mobile phase.
Analytes of interest (and matrix components) remain in the liquid phase or associate only temporarily with the solid stationary phase. The amount of time taken up by the latter is based on characteristics such as charge and polarity of the analytes versus matrix components. A binding-and-wash solvent (different from the elution solvent), provides a relatively crude separation of analytes from the (unwanted) components.

Immunoextraction
Immunoextraction (also known as immunoaffinity purification) is based on the use of antibodies in a solid phase. This separates antibody-bound analytes from free matrix components.

Dilution
When analytes are present in high concentrations, dilution provides a simple and effective methodology to reduce matrix components. Dilution (often called ‘dilute-and-shoot’) is a common method of sample preparation for comprehensive screening and for confirmatory testing for drugs in urine.

Separation
Gas chromatography
GC chromatography uses hydrogen or helium to push molecules into a column (known as the stationary phase). Modifying the column temperature then changes the affinity of molecules in the stationary phase, thereby separating analytes from matrix components (known as the mobile phase). Though largely relevant for volatile, heat-stable compounds, ‘derivatization’ via chemical modification can increase compatibility with GC.
GC mass spectrometry (GC-MS) remains the most common method for comprehensive drug screening in the clinical laboratory.

Liquid chromatography
LC chromatography is largely used for separation of samples before MS analysis. This is largely due to a wide range of LC-compatible analytes and a reduced need for derivatization. The mobile phase in LC uses a combination of organic solvents and water. Adjustments to the ratio between the organics and water redistributes components between the mobile and stationary phases.

Ionization techniques: APCI and electrospray
MS detects charged analytes in the gaseous phase alone. Ionization is required to convert liquid-phase analytes for analysis. The two most common methods in the clinical laboratory consist of atmospheric pressure chemical ionization (APCI) and electrospray ionization.
APCI produces ions by using heat to evaporate the solvent and plasma to ionize the sample. Physical interaction with gaseous analytes leads to formation of negative or positive ions.
Electrospray ionization, on its part, combines electricity, air and heat to produce successively smaller and concentrated droplets from the liquid which elutes off a chromatographic column. This leads to a dramatic increase in charge per unit volume. Ions on the droplet surface desorb from liquid to gas phase, and the latter is introduced into the mass spectrometer.

Sample transfer to mass spectrometer
There are several choices for introducing samples into a mass spectrometer. These range from direct infusion to multidimensional chromatographic separation. The latter enables the staggered delivery of analytes and matrix components. This permits more effective utilization of analyser time by limiting analysis to fractions containing analytes of interest.

Methods of analysis
MS analysis is largely based on quadrupole analysers, time-of-flight (TOF) analysers and tandem mass spectrometers, as well as combinations of the three.

Quadrupole analysers
Linear quadrupole analysers are currently the most common type of mass spectrometer in a clinical laboratory. Called quadrupoles due to the presence of four parallel rods in a square, one pair of (diagonally opposed) rods is positively charged, while the other is negatively charged. The charges are optimized and alternated based on the analyte of specific interest. Via sequential attraction and repulsion, an ion of interest can be programmed to maintain a stable flight path between the rods. The charge and frequency can moreover be rapidly altered to sequentially detect different analytes. Quadrupole analysers have high sensitivity and mass accuracy. On the other hand, they have a limited range in mass/charge (m/z) ratios – which, as noted previously, is a unique identifier for a particular ion.

Time-of-flight analysers
Time-of-flight (TOF) mass spectrometers are based on using an electric field which accelerates gas phase ions to a detector. The time taken for this travel is based on an ion’s m/z ratio, with low m/z ions travelling faster than higher ones.
TOF analysers have an essentially unlimited m/z range and high sensitivity and accuracy, but users face limits in their dynamic range.

Key challenges in clinical MS
Tandem mass spectrometry

Successful identification of an analyte by m/z alone does not always confer specificity. One good example is morphine and hydromorphone. Though the two are distinct, they have identical positive ions, with 286 m/z.
Tandem mass spectrometers (MS/MS) use multiple quadrupoles in series. One typical configuration is to use three quadrupoles. The first and third (denoted Q1 and Q3) use combinations of charge and frequencies as described above (see section on ‘Quadrupole Analysers’). The second quadrupole, denoted q2 (in smaller case), serves as a collision cell with an inert gas (e.g. nitrogen). On entry into q2, ions collide with the inert gas, and fragment into smaller product ions which then pass through Q3 and hit a detector.
In the case of morphine and hydromorphone, q2 entry produces stable product ions (m/z of 153 for the former and 157 for the latter). After this, setting the Q3 charge and frequency to first transmit the product ion for morphine and then change the charge/frequency settings to transmit the  hydromorphone product ion results in a way to measure and differentiate between the two compounds. This approach is also known as multiple reaction monitoring (MRM) and allows a mass spectrometer to scan faster – by targeting specific m/z set points rather than a broad range.

Ion suppression/enhancement and ion ratios
Ion suppression and enhancement are two of the most common problems facing MS. These occur when a substance in a sample interferes with the ionization process of the analytes. These can range from matrix molecules to co-eluting compounds. For example, components in the sample with lower volatility can reduce the efficiency of solvent evaporation, resulting in reduced ion formation.
There are several options to reduce or eliminate such interference, including mobile phase additives to aid ionization.
Another approach is to use ion ratios. When analytes of interest are present alongside structurally similar compounds in complex matrices, interference risks rise due to co-eluting molecules with identical mass. However, ion ratios seek to monitor multiple m/z transitions for each analyte and determine ratios of chromatographic peak area for more abundant fragments to less abundant ones. The use of ion ratios further enhances the specificity of MS.

The future
Endocrinology

Although industry has sought to use antibody-mediated detection to overcome the inherent limitations of immunoassays in identifying proteins and small molecules, these have yet to be meaningfully eradicated.
Rising interest in (regular and more-frequent) testing for vitamin D has also driven implementation of LC-MS/MS (liquid chromatography-tandem mass spectrometry), which separates vitamin D2 from vitamin D3 and provide information on its epimeric form. This is not possible with existing immunoassays.
Meanwhile, although steroid hormone assays for diagnostic and forensic testing continue to grow, a lack of specificity and accuracy at low concentrations has hampered the diagnosis of endocrine disorders. This has led several medical groups to recommend mass spectrometry as the preferred method of analysis, in spite of the high degree of technical competence, skill and experience required to achieve meaningful results.

Metabolomics
Measurements of the genome and proteome need to be accompanied by quantified data on the metabolome to comprehend differences between disease and healthy status, and provide meaningful diagnosis and monitoring of disease. One of the fastest growing areas for MS in metabolomics is the screening of newborns.

Protein analysis

The success of MS in precise measurement of small molecules has driven interest in using it for peptide and protein analysis for diagnostic testing. In spite of some challenges, quantitative proteomics (covering factors such as isotope dilution and m/z transitions) is an especially exciting application for mass spectrometry.

p32 04

Anti-glycolipid antibodies: their role in neurological disease and their detection

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 […]

C281 Euroimmun fig1

Computer-aided immunofluorescence microscopy in autoimmune diagnostics

Indirect immunofluorescence (IIF) is an indispensable method for autoantibody diagnostics, providing high sensitivity and specificity together with a broad antigenic spectrum. However, the microscopic evaluation of the fluorescence patterns is both time-consuming and challenging for laboratory staff, and is, moreover, based on subjective interpretation. Laboratories are increasingly turning to automated systems to facilitate and standardize the IIF readout and interpretation. In recent years various automation systems have been developed, which provide automated digital acquisition of IIF images, discrimination of positive and negative samples, as well as pattern classification for key applications. This article focuses on the EUROPattern system, which provides computer-aided immunofluorescence microscopy for anti-nuclear antibodies (ANA), anti-neutrophil granulocyte cytoplasm antibodies (ANCA), antibodies against double-stranded DNA (anti-dsDNA) on Crithidia luciliae, monospecific antigen microdots (EUROPLUS) and transfected cell-based assays e.g. for anti-neuronal antibodies. The accuracy of automated evaluation compared to visual assessment has been investigated in various published studies.

by Dr Jacqueline Gosink

ANA
ANA represent a key diagnostic criterion for many autoimmune diseases, including systemic lupus erythematosus (SLE), mixed connective tissue disease, Sjögren’s syndrome, systemic sclerosis, polymyositis, dermatomyositis and primary biliary cirrhosis. The gold standard for ANA determination is IIF on human epithelial (HEp-2) cells. This substrate provides the complete antigen spectrum and allows investigation of over 100 different autoantibodies. Observation of the fluorescence pattern enables classification of the antibody or antibodies present in the patient sample. Positive results are confirmed by monospecific tests such as ELISA, immunoblot or IIF microdot assays.

The automated evaluation of HEp-2 cells includes reliable discrimination of positive and negative ANA results, as well as classification of all ANA patterns [1] (Figure 1), encompassing homogeneous, speckled, nuclear dots, nucleolar, centromeres, nuclear envelope and cytoplasmic. The ANA patterns identified by EUROPattern correspond to the competent level reporting defined by the International Consensus on ANA Patterns (ICAP; www.anapatterns.org). Mixed patterns, which occur when more than one antibody is present, are also recognized and reported as such. The pattern is assigned by analysing its features and comparing it to a reference database of over 5000 images, corresponding to 115,000 cells. Unspecific signals originating from outside of the cells are identified by means of a DNA counterstain and subsequently rejected. The evaluation also includes titre designations with confidence values for the detected antibodies. Results from the HEp-2 screening can be monospecifically confirmed using microdot substrates of purified antigens, which are incubated and evaluated in parallel.

To assess the diagnostic accuracy, the automated evaluation was compared to conventional visual interpretation by experts in the field using 351 patient sera [2]. The concordance for positive/negative discrimination was 99%, with an analytical sensitivity of 100% and a specificity of 98%. In 60% of samples, the pattern, including variable mixed patterns, was recognized completely by the software. In 94% of samples, the main pattern was correctly designated. A further study showed 79% correct pattern assignment.

Anti-dsDNA antibodies
Anti-dsDNA antibodies are a hallmark of SLE and represent an important criterion for diagnosis. Their prevalence in SLE ranges from 30% to 98% in different studies, depending among other things on the test method used. Like the gold standard Farr assay, IIF using Crithidia luciliae as the substrate (CLIFT) is considered to have a very high disease specificity. The method takes advantage of the kinetoplast of C. luciliae, which is rich in DNA but contains hardly any other antigens, thus enabling highly selective detection of anti-dsDNA antibodies. However, manual reading of the fluorescence signals is subjective and leads to high intra- and inter-laboratory variation, making standardized automated evaluation a desirable goal.
Automated interpretation of CLIFT has recently been incorporated into the EUROPattern system [3] (Figure 2). The software is able to recognize the organelles of the protozoan and evaluates the specific kinetoplast fluorescence rather than just dark-light classification, increasing the reliability of the evaluation. Results are classified as positive or negative, and include a titre designation based on the fluorescence intensity.

In a clinical study, automated and visual evaluation of C. luciliae IIF was compared using 569 consecutive sera submitted for routine anti-dsDNA screening and 100 sera from healthy blood donors. The automated system recognized all 73 of the anti-dsDNA positive samples identified by the visual evaluation. Moreover, 93% of the titre designations were concordant. The overall sensitivity of the system amounted to 100% with a high specificity of 97%. Compared to visual microscopy the overall accuracy was 97%.

ANCA
ANCA are important serological markers for diagnosis and differentiation of autoimmune vasculitides, especially granulomatosis with polyangiitis (GPA, formally known as Wegener’s granulomatosis), which is characterized by autoantibodies against proteinase 3 (PR3), and microscopic polyangiitis, which is typified by autoantibodies against myeloperoxidase (MPO). In addition, ANCA can be found in chronic inflammatory bowel diseases. ANCA are detected by IIF with monospecific confirmation using ELISA, immunoblot or IIF microdot assays.
The IIF substrates ethanol-fixed and formalin-fixed granulocytes are used to identify the typical ANCA staining patterns of anti-PR3 (cytoplasmic, cANCA) and anti-MPO (perinuclear, pANCA) antibodies. An additional substrate consisting of HEp-2 cells coated with granulocytes allows immediate differentiation between ANCA and ANA, while purified antigen microdots of PR3, MPO or glomerular basement membrane (GBM) antigen provide simultaneous monospecific antibody characterization. The different substrates are incubated and automatically evaluated in parallel as BIOCHIP mosaics, thus providing ANCA screening and confirmation in one step.

Evaluation software such as EUROPattern provides automated positive/negative discrimination of samples, as well as recognition of pANCA and cANCA patterns [1] (Figure 3). Further pattern constellations such as DNA-ANCA (atypical pANCA, xANCA), which can arise from antibodies against lactoferrin or other antigens, are also taken into account by the software. The automated system proposes a result based on the recognized cellular patterns and the results on the antigen microdots. An estimated titre with a confidence value is given.

Anti-neuronal antibodies
Neuronal cell-surface autoantibodies occur in autoimmune encephalitis and their detection can secure an early diagnosis, enabling immediate treatment which is critical for patient outcome. In recent years a considerable number of novel target antigens has been discovered, for example, glutamate receptors of type NMDA and AMPA, GABAB receptors, voltage-gated potassium channel-associated proteins LGI1 and CASPR2, DPPX and IgLON5.

Diagnostic tests for the new parameters are based on recombinant-cell (RC) IIF, in which transfected cells expressing the relevant antigen are used for monospecific antibody detection. This test method enables authentic presentation of the fragile membrane-associated surface antigens. Since many of the autoantibody markers are rare and do not always overlap, a multiparametric screening using BIOCHIP mosaics made up of different substrates is recommended. Results for RC-IIF assays can be evaluated automatically using a newly developed function of EUROPattern. The system automatically takes digital images of the substrates and provides a positive/negative classification.
The quality of the acquired images was assessed by comparing on-screen appraisal with visual microscopy using 753 incubations of numerous serum samples sent to a clinical immunology laboratory [4]. Ambiguous fluorescence signals detected at the microscope were excluded to avoid inter-reader deviations. The two evaluation strategies revealed a concordance of 100% with respect to positive/negative discrimination, confirming the high quality of the images.

Arbovirus antibodies
Immunofluorescence microscopy is also useful for infectious disease diagnostics. For example, infections with Zika virus, dengue virus and chikungunya virus are difficult to tell apart clinically as they manifest with similar symptoms and are endemic in much the same regions. Serological tests are an important diagnostic method, especially beyond the short viremic phase when direct detection is no longer effective. Viral antibodies can be detected by IIF using virus-infected cells. However, cross reactions between flavivirus antibodies can occur.

A BIOCHIP mosaic comprising substrates for Zika, dengue and chikungunya viruses enables parallel antibody determination, aiding clarification of cross reactivities and supporting differential diagnosis. The substrates can be evaluated semi-automatically using the digital image acquisition function of EUROPattern. In particular, inspection of the images side-by-side on the computer screen considerably facilitates the interpretation.

Fully automated immunofluorescence microscopy
Computer-aided fluorescence microscopy can be further standardized and facilitated through use of complementary hardware. The EUROPattern microscope (Figure 4) has been tailored to the requirements of immunofluorescence. Next to the high-precision optical system, it has a controlled LED, which maintains a constant light flux, ensuring highly reproducible results. The cLED has an extremely long life span without maintenance (over 50,000 hours) and low power consumption, ensuring cost-effectiveness for laboratories. The microscope is equipped with a slide magazine which can process up to 500 analyses in succession within 2.5 hours (18 seconds per field), correctly identifying the slides by means of matrix codes.

Results from the automated IIF evaluation can be viewed and validated directly at the computer screen, enabling a diagnosis to be established quickly and efficiently. The high-resolution images are sharply focused with the aid of a counterstain. The counterstain also serves to verify correct performance of the incubation. Negative results can be verified in batches, while positive samples can be individually checked and confirmed by the medical technologist. Results from different serum dilutions and substrates are consolidated into one report per patient, and new findings are compared with previous records. Final results can be signed electronically and forwarded at a click.

Perspectives
The need for standardization and automation in IIF is tremendous in all fields of autoimmune diagnostics. In particular, manual evaluation of results is time consuming and subjective. Automation platforms with harmonized software and hardware components have in recent years contributed enormously to the standardization and simplification of the evaluation process, especially for ANA, ANCA and CLIFT. Advanced software provides positive/negative classification, pattern recognition and titre designation at a quality equivalent to visual microscopy. The recording of tissue substrates, such as liver, kidney, stomach, esophagus, small intestine, heart and neuronal tissue, is also feasible. Future development will focus on the recognition of organ- and non-organ-specific autoantibodies on tissues, for example antibodies against mitochondria, epithelial membrane, epidermal basement membrane, desmosomes, heart muscle and neuronal antigens. The continued development of automated evaluation systems is anticipated to lead to even greater standardization of IIF and further reductions in workflow for diagnostic laboratories.

References
1. Krause et al. Lupus 2015: 24: 516-29
2. Voigt et al. Clin. Devel. Immunol. 2012: vol 2012, article ID 651058
3. Gerlach et al. J. Immunol. Res. 2015: vol 2015, article ID 742402
4. Fraune et al. Autoimmunity Reviews 15 (2016) 937-942

The author
Jacqueline Gosink, PhD
EUROIMMUN AG, Seekamp 31, 23560 Luebeck, Germany
E-mail: j.gosink@euroimmun.de

p38 02

Autoimmune diagnostics by immuno- fluorescence: variability and harmonization

by Dr Petraki Munujos The antinuclear antibodies (ANA) determination is one of the most commonly used techniques in the autoimmunity clinical laboratory. Far from being outdated, indirect immunofluorescence (IF) is a powerful laboratory tool not only for clinical diagnostics, but for disease follow-up and prognosis estimation as well. Unlike other more precise quantitative techniques, IF […]

C282 E77 UriSed3

Evaluation of UriSed 3 automated urine microscopy analyser

Urinalysis may provide evidence of significant renal disease in asymptomatic patients. The microscopic urinalysis is vital to making diagnoses in many asymptomatic cases, including urinary tract infection, urinary tract tumors, occult glomerulonephritis, and interstitial nephritis.

Presence or absence of different particles in urine sediment is crucial for clinical decision making. Urine sediment cells or particles provide important information for the diagnosis of renal or urinary diseases [1]. The patented UriSed Technology was developed to reduce the shortcomings of manual microscopy through automation. [2]. The UriSed analysers provide a reliable and reproducible solution since 2007 [3]. The new generation instrument based on the improved UriSed Technology, UriSed 3 was introduced in the market in 2015. UriSed 3 is an automated urine microscopy analyser with a revolutionary particle visualization utilizing both bright-field and phase-contrast microscopy. In the present study, we evaluated the analytical performance of UriSed 3 Automated Urine Microscopy Analyser (Manufactured by 77 Elektronika Kft., Budapest) and compared the results to those from manual microscopy using standardized KOVA counting chambers.

UriSed 3 provides quantitative Red Blood Cell (RBC) and White Blood Cell (WBC) results, and semi-quantitative results for all other particle types: WBC Clumps (WBCc), Squamous Epithelial Cells (EPI), Non-squamous Epithelial Cells (renal tubular and urothelium cells) (NEC), Crystals (CRY): Calcium oxalate dihydrate (CaOxd), Calcium oxalate monohydrate (CaOxm), Uric acid (URI), and Triple-phosphate crystals (TRI), Hyaline casts (HYA), Pathological casts (PAT), Bacteria (cocci-like and rod-like) (BACc, BACr), Yeasts (YEA), Spermatozoa (SPRM) and Mucus (MUC) [4].

Phase-contrast microscopy by UriSed 3
Phase-contrast microscopy is an optical microscopy technique that converts phase shifts in light passing through a transparent specimen to brightness changes in the image. Phase shifts themselves are invisible, but become visible when shown as brightness variations. In particular, for urinary sediment examination, phase-contrast supplies an optimal identification of particles with a low refractive index (e.g., hyaline casts and RBC devoid of their hemoglobin content, the so-called “ghost RBC”) and of cellular morphological details) [9, 10]. Therefore the use of phase-contrast microscopy is encouraged also by international guidelines on urinalysis [5, 6].

The measurement technique of the UriSed 3 instrument is a combination of a bright-field microscope and a phase-contrast microscope in one optical system. Preparation of the UriSed 3 analyser for measurement takes only a few minutes. It needs distilled water for washing its pipette, and patented disposable plastic cuvettes for sample investigation. The instrument throughput is up to 120 samples per hour. The whole measurement process is completely automatic: 200 µl of urine sample is dispensed into the cuvette, then spinning the cuvette for a few seconds gently deposits formed elements into a monolayer at the bottom of the cuvette. The built-in digital camera takes and saves both a bright-field and a phase-contrast microscopic image from the same view-field at 15 different positions of the sediment layer. Information from both whole view-field images are evaluated by a neural network based image processing software.

Material and methods

Analysis of 311 samples was performed to evaluate UriSed 3 analytical performance compared to the manual microscopy urine examination method. Both measurements were carried out with the same anonymous urine samples. Fresh, native urine samples were used, that were typically held for no more than 4 hours before being analysed, as recommended in the relevant guidelines [5,6] to prevent change in the morphology of the particles. Samples were mixed until homogeneous and then split and run on each measuring procedure as close to the same time as possible. The standardized microscopic urinalysis of native samples (Level 3) was followed by using a KOVA counting chamber. The particle concentration for all particle types was evenly distributed in the evaluated urine samples. Carry-over, precision, diagnostic tests such as sensitivity, specificity, diagnostic accuracy, concordance and one category concordance were investigated according to well-established protocols [7].

Results
No carry-over was detected in any of the samples. UriSed 3 has better precision than microscopy at all of the tested RBC and WBC concentrations. The majority of all coefficients of variation obtained for within series imprecision (CV) using UriSed 3 was 7-16% versus 5,5-67% in case of manual microscopy [8]. Good correlation can be found between UriSed 3 and manual counting chamber for formed elements. The Pearson correlation of quantitative parameters are 0.91 (RBC), 0.93 (WBC). The clinical evaluation of UriSed 3 was based on McNemar test and concordance study. The results are shown in the table above.

Conclusion
UriSed 3 instruments utilize phase-contrast and bright-field microscopy to combine original and innovative technologies whose aim is the progressive improvement of automated urinary sediment examination and the progressive approach to the gold standard manual microscopy method. The automated measurement process of UriSed 3 is reproducible and operator-independent. Those sediment particles that are mostly transparent become visible with phase-contrast microscopy by UriSed 3, which is a spectacular advantage and leads to specific improvement in recognition at several particle types.

References
1. Fogazzi GB, The Urinary Sediment an Integrated View Third Edition. Milano: Elsevier, 2010.
2. Barta Z, Kránicz T, Bayer G. UriSed Technology – A Standardised Automatic Method of Urine Sediment Analysis. European Infectious Disease 2011;5:139–42.
3. Zaman Z, Fogazzi GB, Garigali G, Croci MD, Bayer G, Kránicz T. Urine sediment analysis: analytical and diagnostic performances of sediMAX – a new automated microscopy image-based urine sediment analyser. Clin Chim Acta 2010; 411: 147-154.
4. Fogazzi GB, Garigali G. The Urinary Sediment by UriSed Technology. A New Approach to Urinary Sediment Examination. Milano: Elsevier, 2013.
5. Kouri T, Fogazzi G, Hallander H, Hofmann W, Guder WG, editors. European Urinalysis Guidelines. Scand J Clin Lab Invest 2000; 60 (Suppl 231): 1-96.
6. Clinical and Laboratory Standard Institute (ex NCCLS). Document GP16-A3 – Urinalysis; Approved guideline, 3rd ed. Wayne, PA: CLSI, 2009.
7. T. Kouri, A. Gyory, R.M. Rowan. ISLH Recommended Reference Procedure for the enumeration of Particles in Urine. Laboratory Hematology 9:58-63, 2003.
8. Haber MH, Galagan K, Blomberg D, Glassy EF, Ward PCJ, editors. Color Atlas of Urinary Sediment; An Illustrated Field Guide Based on Proficiency Testing. Chicago: CAP Press, 2010.
9. Brody L, Webster MC, Kark RM. Identification of elements of urinary sediment with phase-contrast microscopy. JAMA 1968; 206: 1777-1781.
10. Spencer E. and Pedersen Ib. Hand Atlas of the Urinary Sediment. Bright-field, Phase-Contrast, and Polarized Light. Copenhagen: Munksgaard, 1971.

More information on UriSed 3 is available from the manufacturer:
77 Elektronika Kft., Budapest, HUNGARY
Email: sales@e77.hu, web: en.e77.hu

The author
Erzsébet Nagy MD,
Honorary Associate Professor
Head Phisician of Central Laboratory; Hospitaller Brothers of St. John of God Hospital, Budapest

greiner bio one advertorial CLI Nov

Small tubes, great impact

Scientific Lit picture 02

SCIENTIFIC LITERATURE REVIEW: Colorectal cancer

Highly sensitive stool DNA testing of Fusobacterium nucleatum as a marker for detection of colorectal tumours in a Japanese population

Suehiro Y, Sakai K, Nishioka M, Hashimoto S, Takami T, Higaki S, Shindo Y, Hazama S, Oka M, Nagano H, Sakaida I, Yamasaki T. Ann Clin Biochem. 2016; pii: 0004563216643970. [Epub ahead of print]

BACKGROUND: Accumulating evidence shows an over-abundance of Fusobacterium nucleatum in colorectal tumour tissues. Although stool DNA testing of Fusobacterium nucleatum might be a potential marker for the detection of colorectal tumours, the difficulty in detecting Fusobacterium nucleatum in stool by conventional methods prevented further explorations. Therefore, we developed a droplet digital polymerase chain reaction (PCR) assay for detecting Fusobacterium nucleatum in stool and investigated its clinical utility in the management of colorectal tumours in a Japanese population.
METHODS: Feces were collected from 60 healthy subjects (control group) and from 11 patients with colorectal non-advanced adenomas (non-advanced adenoma group), 19 patients with colorectal advanced adenoma/carcinoma in situ (advanced adenoma/carcinoma in situ (CIS) group) and 158 patients with colorectal cancer of stages I to IV (colorectal cancer group). Absolute copy numbers of Fusobacterium nucleatum were measured by droplet digital PCR.
RESULTS: The median copy number of Fusobacterium nucleatum was 17.5 in the control group, 311 in the non-advanced adenoma group, 122 in the advanced adenoma/CIS group, and 317 in the colorectal cancer group. In comparison with that in the control group, the Fusobacterium nucleatum level was significantly higher in the non-advanced adenoma group, the advanced adenoma/CIS group and the colorectal cancer group.
CONCLUSIONS: This study illustrates the potential of stool DNA testing of Fusobacterium nucleatum by droplet digital PCR to detect individuals with colorectal tumours in a Japanese population.

A genome-wide assessment of variations of primary colorectal cancer maintained in metastases

Cai Z, Han S, Li Z, He L, Zhou J, Huang W, Xu Y. Gene 2016; 595(1): 18–24.

Colorectal cancer (CRC) is a highly heterogeneous disease that is the third leading cause of cancer-related deaths worldwide. This study presents a genome-wide assessment of variations in primary colorectal cancer maintained in metastases, even in distant metastases. The purpose of this study was to determine whether intratumor heterogeneity is related to disease progression and metastasis in CRC. The results showed that 882 single nucleotide polymorphism (SNP) associated genes and 473 copy number variant (CNV) associated genes specific to metastasis were found. In addition, 57 SNPs mapped to miRNAs showed significant differences between primary tumours and metastases. Functional annotation of metastasis-specific genes suggested that adhesion and immune regulation may be essential in the development of tumours. Moreover, the locus rs12881063 in the fourteenth chromosome was found to have a high rate of the G/C type in metastases. The rate of the G/C type in nearby lymph node metastases was 66.7%, while the rate of the G/C type in distance lymph node metastases was 83.3%. These results indicate that rs12881063 may be the basis for clinical diagnosis of CRC metastasis.

High tumour mast cell density is associated with longer survival of colon cancer patients

Mehdawi L, Osman J, Topi G, Sjölander A. Acta Oncol. 2016; 55(12): 1434–1442.

BACKGROUND: Inflammatory cells and inflammatory mediators play an important role in colorectal cancer (CRC). Previous studies have shown that CRC patients with increased expression of cysteinyl leukotriene receptor 1 (CysLTR1) have a poorer prognosis, and Cysltr1-/- mice display fewer intestinal polyps. However, the role of mast cells (MCs) in colon cancer progression remains unclear. The aim of the present study was to explore the relevance of MCs in CRC.
MATERIAL AND METHODS: A tissue microarray from 72 CRC patients was stained with MC anti-tryptase and -chymase antibodies. Mouse colon tissue was stained with MC anti-tryptase antibody. Immunohistochemistry was used to identify MCs in patients and mice.
RESULTS: Patient colon cancer tissue had in comparison with normal colon tissue a reduced number of MCs, predominantly of chymase-positive cells. Further analysis revealed that patients with a relative high MCD in their cancer tissues showed significantly longer overall survival compared to those with a low MCD [hazard ratio (HR) 0.539; 95% confidence interval (CI), 0.302–0.961]. Similar results were observed in subgroups of patients with either no distant metastasis (p = 0.004), or <75 years (p = 0.015) at time of diagnosis. Multivariate Cox analysis showed that MCD independently correlated with reduced risk of death in colon cancer patients (HR 0.380; 95% CI 0.202-0.713). Additionally, a negative correlation was found between cytoplasmic CysLTR1 expression and number of MCs. In agreement, in the CAC mouse model, Cysltr1-/- mice showed significantly higher MCs in their polyp/tumor areas compared with wild-type mice.
CONCLUSION: A high MCD in cancer tissue correlated with longer patient survival independently from other risk factors for CRC. The concept that MCs have an anti-tumor effect in CRC is further supported by the findings of a negative correlation with CysLTR1 expression in patients and a high MCD in colon polyps/tumors from CysLTR1-/- mice.

Are hemorrhoids associated with false-positive fecal immunochemical test results?

Kim NH, Park JH, Park DI, Sohn CI, Choi K, Jung YS. Yonsei Med J. 2016; 58(1):150–157.

PURPOSE: False-positive (FP) results of fecal immunochemical tests (FITs) conducted in colorectal cancer (CRC) screening could lead to performing unnecessary colonoscopies. Hemorrhoids are a possible cause of FP FIT results; however, studies on this topic are extremely rare. We investigated whether hemorrhoids are associated with FP FIT results.
MATERIALS AND METHODS: A retrospective study was conducted at a university hospital in Korea from June 2013 to May 2015. Of the 34,547 individuals who underwent FITs, 3946 aged ≥50 years who underwent colonoscopies were analysed. Logistic regression analysis was performed to determine factors associated with FP FIT results.
RESULTS: Among 3946 participants, 704 (17.8%) showed positive FIT results and 1303 (33.0%) had hemorrhoids. Of the 704 participants with positive FIT results, 165 had advanced colorectal neoplasia (ACRN) and 539 had no ACRN (FP results). Of the 1303 participants with hemorrhoids, 291 showed FP results, of whom 81 showed FP results because of hemorrhoids only. Participants with hemorrhoids had a higher rate of FP results than those without hemorrhoids (291/1176, 24.7% vs. 248/2361, 10.5%; p<0.001). Additionally, the participants with hemorrhoids as the only abnormality had a higher rate of FP results than those experiencing no such abnormalities (81/531, 15.3% vs. 38/1173, 3.2%; p<0.001). In multivariate analysis, the presence of hemorrhoids was identified as an independent predictor of FP results (adjusted odds ratio, 2.76; 95% confidence interval, 2.24-3.40; p<0.001).
CONCLUSION: Hemorrhoids are significantly associated with FP FIT results. Their presence seemed to be a non-negligible contributor of FP results in FIT-based CRC screening programmes.