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.
Molecular testing is increasingly recognized for guiding patient management and development of new targeted therapies. Meanwhile, there is an increased demand to perform testing on smaller volumes of tissue. Recent literature reports the use of ultrasensitive techniques to detect DNA mutations and translocations from stained cytology smears [1].
by Dr Laleh Hakima, Dr Maja H. Oktay and Prof. Sumanta Goswami
Introduction
Mutational analyses are crucial for guiding treatment decisions. This is particularly important for targeted therapies with tyrosine kinase inhibitors (TKI) for lung non-small cell carcinoma (NSCC) and for surgical management of thyroid nodules with indeterminate cytological diagnoses. Both lung and thyroid lesions are frequently diagnosed using fine need aspiration (FNA). FNA is a preferred method of obtaining diagnostic samples compared to surgical excisions or core biopsies. The FNA procedure is minimally invasive, rapid, cost-effective and has reduced procedure-related complications.
Molecular diagnostic tests of cytological samples are often performed using cytology cell block preparations. However, insufficient cellularity of cell blocks is a frequent limitation to these tests. In addition, errors introduced by formalin fixation may also interfere with accurate detection of mutations. Recent studies have reported successful testing using cytology smears stained with Diff-Quik®, which are air dried and fixed in alcohol. This approach has several advantages compared to testing on paraffin-embedded tissue (PET). Alcohol is a great preservative for DNA and RNA. The sample quality is superior to PET because FNA samples are typically enriched for cancer cells and cytology smears yield intact whole nuclei rather than the nuclear fragments that are obtained by cutting PET [2]. Lastly, testing performed directly on microdissected tumour cells ensures that material is sufficient and representative of the tumour and results in a faster turnaround time. We used cytological direct smears stained with Diff-Quik® and Papanicolaou (Pap) to detect commonly encountered mutations in non-small cell lung carcinoma and thyroid carcinoma [1].
Lung- and thyroid-carcinoma mutations and fusion gene testing targets
Approximately 64% of all lung adenocarcinomas harbour somatic driver mutations. According to The Lung Cancer Mutation Consortium, the frequency of EGFR and KRAS mutations are 23% and 25%, respectively. The incidence of EML4–ALK translocation, mainly detected in non-smoker patients with wild-type EGFR and KRAS genes, is approximately 6% [2, 3]. Other less frequent mutations include: BRAF ~3%, PIK3CA ~3%, MET amplifications ~2%, ERBB2 (HER2/NEU) ~1%, MAP2K1 ~0.4%, and NRAS ~0.2% [2]. Additionally, new driver mutations have recently been identified in lung cancer patients [3]. The most common mutations in thyroid carcinoma involve BRAF, KRAS, and RET/PTC genes. The mutations in BRAF and KRAS are point mutations, whereas RET (PTC) mutations are gene rearrangements that result in fusion of the tyrosine kinase domain of the RET gene to various unrelated genes [4–7].
The EGFR mutation status of the cancer is associated with its responsiveness or resistance to EGFR TKI therapy. The EGFR gene is located on chromosome 7p11.2, spans about 200kb, and contains 28 exons. The gene encodes a protein of 464 amino acids. The EGFR protein is composed of an N-terminal extracellular ligand-binding domain, a transmembrane lipophilic segment, and a C-terminal intracellular region containing a tyrosine kinase domain. The EGFR tyrosine kinase modulates cell proliferation and survival. Activation of EGFR initiates signalling cascades involving several downstream pathways, including Ras GTPases, which induce crucial cellular responses, such as proliferation, differentiation, motility, and survival. EGFR mutations associated with objective responses to single-agent TKI therapy in lung adenocarcinomas are preferentially observed in females of East Asian ethnicity who are never smokers and have adenocarcinoma with lepidic growth pattern (formerly bronchioloalveolar carcinoma). In adenocarcinomas, the majority of mutations have been identified in exons 18–21 of the EGFR gene. These mutations can be roughly classified into three major categories: in-frame deletions in exon 19, insertion mutations in exon 20, and missense mutations in exons 18–21 [8].
The fusion of the echinoderm microtubule-associated protein-like 4 (EML4) gene to the anaplastic lymphoma kinase (ALK) gene, EML4–ALK, is the most common fusion and results from the joining of exons 1–13 of EML4 to exons 20–29 of ALK. At least seven EML4–ALK variants (V1–V7) have been identified in lung adenocarcinomas. All seven variants are formed through the fusion of the intracellular tyrosine kinase domain of ALK with a variably truncated EML4 gene promoter. Activated ALK is involved in the inhibition of apoptosis and the promotion of cellular proliferation through activation of downstream PI3K/AKT1- and MAPK1-signalling pathways. The key downstream effectors on the ALK pathway include the Ras-activated protein, mitogen-activated protein kinase 1 [MAPK1; also known as extracellular signal regulated kinase (ERK)], phosphatidylinositol 3-kinase (PI3K), and signal transducer and activator of transcription 3 (STAT3) signalling pathways. Ras/MAPKK1/MAPK1 pathways are critical for cell proliferation, whereas the PI3K/AKT1 and STAT3 pathways are important for cell survival. The histology of these tumours is typically characterized by mucin production and either a solid growth pattern containing signet ring cells in Western patients or an acinar growth pattern in Asian patients. Compared with patients with wild-type ALK and EGFR, patients with the EML4–ALK fusion gene tend to be younger, of Asian ethnicity, diagnosed at an advanced clinical stage at presentation, male dominant, and more likely to be never smokers, but with a comparable response rate to chemotherapy and overall survival. The EML4–ALK fusion gene is typically detected by fluorescence in-situ hybridization (FISH). It has been reported that although ALK-fusion-positive lung cancers are resistant to the EGFR TKIs, gefitinib, and erlotinib, they are sensitive to small molecule TKIs against ALK [8].
The BRAFV600E point mutation involves nucleotide 1799 and results in a valine-to-glutamate substitution at residue 600 (V600E). B-Raf is a serine/theronine protein kinase involved in MAPK/ ERK signalling pathway and in regulation of cell proliferation and differentiation. It is found in approximately 40–45% of papillary thyroid carcinomas. Not all variants are equally affected; 60% of classic papillary, 80% of tall cell variant, and 10% of follicular variant harbour this mutation. Its detection is clinically significant because if represents a prognostic marker for thyroid papillary carcinoma, it is associated with extrathyroidal extension, advanced tumour stage at presentation, and lymph node or distant metastases. BRAFV600E point mutation is also an independent predicator of treatment failure and tumour recurrence, even with patients with low-stage disease [4].
Case selection and molecular analysis techniques
Thirty-one cases of lung adenocarcinomas and 26 thyroid carcinomas (17 classic papillary, 7 follicular variant, and 2 follicular carcinomas) were selected from the archives. Molecular analysis was performed on PET and from either Pap or Diff-Quik® stained smears for each case. The following mutations in lung adenocarcinomas were tested: EGFR point mutations in exons 20 and 21, in-frame deletions in exon 19; KRAS point mutations in codons 12, 13, and 61; and EML4–ALK translocation. Thyroid carcinomas were tested for the BRAFV600E point mutation.
Smears were reviewed by two cytopathologists and areas containing at least 50 cancer cells without necrosis or inflammation were marked for analysis [9, 10]. Tumour cells from marked areas were microdissected using RNA/DNA co-purification solution (Zymo Technologies) [5, 6]. QClamp xenonucleic acid (XNA) technology was used to detect mutations in EGFR, KRAS and BRAF genes. The qClamp, a wild-type sequence-specific XNA probe, has a melting point higher than 72 °C and remains attached to the wild-type DNA during the PCR extension stage. If there is a mutation, the probe dissociates from the template, which allows amplification. The technology was optimized using wild-type DNA (Promega, Madison, WI) resulting in a DNA shift of more than seven cycle thresholds (CTs) with the XNA clamp. In samples with mutations, the shift was less than 5 CTs (Fig. 1).
Quantitative RT-PCR (RT-qPCR) was used to detect EML–ALK translocations. PCR primers were used to amplify both the 3ʹ and 5ʹ ends of the ALK transcript (Aanera Biotech). In the presence of the translocation, the transcription of the EML4–ALK fusion gene (under the control of a stronger EML promoter) resulted in higher than expected 3ʹ ends than 5ʹ ends which lead to lower CT values for the 3ʹ transcript. A difference of more than 10 CTs between 3ʹ and 5ʹ ends was considered positive (Fig. 1).
The sensitivity of our assay was determined using two lung cancer cell lines, H1975 and H2228 (ATCC), harbouring the L858R mutation in the EGFR gene and the EML4–ALK translocation, respectively. The mutation detection rate was 1%.
Results
Approximately 80.6% of lung cases had some form of molecular alteration detected. There was 100% concordance between PET and cytology smears. All cases tested positive by our laboratory were also positive by the reference laboratory. However, seven cases that tested negative for EGFR mutation by the reference laboratory were found to be positive in our lab. In other words, clamp qPCR methodology detected approximately 20% more EGFR mutations than the reference laboratory. Likewise, three cases that were negative for KRAS by the reference laboratory, tested positive in our laboratory. In addition, there were six cases (19%) that had more than one molecular alteration in our cohort. Five cases had two mutations (three had EGFR exon 19 and KRAS codon 12 mutations, two had KRAS codon 12 and EML4–ALK mutations) and one case had three mutations (EGFR exon 19 and 21 and KRAS codon 12 mutation). Although EGFR and KRAS mutations were previously thought to be mutually exclusive, our results confirm recent reports of simultaneous mutations detected in study samples [7]. Given the known heterogeneity of cancers this finding may be expected, but might not have been detected previously because most analyses are performed using standard-sensitivity techniques.
Of 26 thyroid cases, 18 (81%) were positive for BRAFV600E mutation on both PET and cytology smears. Concordant results were obtained from both cytology smears and PET from cases tested by us and the reference laboratory. Both follicular carcinoma cases tested negative on cytology smears and PET for BRAFV600E. The percentage of patients with a BRAF mutation detected by us was higher than expected from the literature (81% versus 50%) reflecting our ultrasensitive approach to mutation detection [10].
Conclusion
Our results indicate that qClamp technology and the RT-qPCR approach can be used successfully to detect most common targetable molecular alterations from cytology smears of lung and thyroid carcinomas. We report successful DNA and RNA isolation from both Diff-Quik® and Pap stained smears. This methodology represents an ultrasensitive and accurate approach with a 1% mutation detection rate and a decreased turnaround time of 1 to 2 days. Such an ultrasensitive method for molecular testing is essential as smaller amounts of diagnostic material become available and targeted approaches will be aimed at both molecular alterations present in the majority of cells, and at those present in a minority of cells that potentially may represent a subpopulation susceptible to recurrence [11].
Abbreviations
Table 1 shows the gene symbols and the names and symbols of the proteins encoded by those genes.
References
1. Oktay MH, Adler E, Hakima L, Grunblatt E, Pieri E, Seymour A, Khader S, Cajigas A, Suhrland M, Goswami S. The application of molecular diagnostics to stained cytology smears. J Mol Diagn. 2016; 18(3): 407–415.
2. Tsao MS, Sakurada A, Cutz JC, Zhu CQ, Kamel-Reid S, Squire J, Lorimer I, Zhang T, Liu N, Daneshmand M, Marrano P, da Cunha Santos G, Lagarde A, Richardson F, Seymour L, Whitehead M, Ding K, Pater J, Shepherd FA. Erlotinib in lung cancer – molecular and clinical predictors of outcome. N Engl J Med. 2005; 353: 133–144.
3. Campbell JD, Alexandrov A, Kim J, Wala J, Berger AH, Pedamallu CS, Shukla SA, Guo G, Brooks AN, Murray BA, Imielinski M, Hu X, Ling S, Akbani R, Rosenberg M, Cibulskis C, Ramachandran A, Collisson EA, Kwiatkowski. DJ, Lawrence MS, Weinstein JN, Verhaak RG, Wu CJ, Hammerman PS, Cherniack AD, Getz G, Cancer Genome Atlas Research Network, Artyomov MN, Schreiber R, Govindan R, Meyerson M. Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas. Nat Genet. 2016; 48(6): 607–616.
4. Nikiforov YE. Molecular diagnostics of thyroid tumors. Arch Pathol Lab Med. 2011; 135(5): 569–577.
5. Gupta N, Dasyam AK, Carty SE, Nikiforova MN, Ohori NP, Armstrong M, Yip L, LeBeau SO, McCoy KL, Coyne C, Stang MT, Johnson J, Ferris RL, Seethala R, Nikiforov YE, Hodak SP. RAS mutations in thyroid FNA specimens are highly predictive of predominantly low-risk follicular-pattern cancers. J Clin Endocrinol Metab. 2013; 98(5): 914–922.
6. Nikiforov YE, Ohori NP, Hodak SP, Carty SE, LeBeau SO, Ferris RL, Yip L, Seethala RR, Tublin ME, Stang MT, Coyne C, Johnson JT, Stewart AF, Nikiforova MN. Impact of mutational testing on the diagnosis and management of patients with cytologically indeterminate thyroid nodules: a prospective analysis of 1056 FNA samples. J Clin Endocrinol Metab. 2011; 96(11): 3390–3397.
7. Nikiforov YE, Yip L, Nikiforova MN. New strategies in diagnosing cancer in thyroid nodules: impact of molecular markers. Clin Cancer Res. 2013; 19(9):2283–2288.
8. Cheng L, Alexander RE, Maclennan GT, Cummings OW, Montironi R, Lopez-Beltran A, Cramer HM, Davidson DD, Zhang S. Molecular pathology of lung cancer: key to personalized medicine. Mod Pathol. 2012; 25: 347–369.
9. Marchetti A, Milella M, Felicioni L, Cappuzzo F, Irtelli L, Del Grammastro M, Sciarrotta M, Malatesta S, Nuzzo C, Finocchiaro G, Perrucci B, Carlone D, Gelibter AJ, Ceribelli A, Mezzetti A, Iacobelli S, Cognetti F, Buttitta F. Clinical implications of KRAS mutations in lung cancer patients treated with tyrosine kinase inhibitors: an important role for mutations in minor clones. Neoplasia. 2009; 11: 1084–1092.
10. Russo M, Malandrino P, Nicolosi ML, Manusia M, Marturano I, Trovato MA, Pellegriti G, Frasca F, Vigneri R. The BRAF (V600E) mutation influences the short and medium-term outcomes of classic papillary thyroid cancer, but is not an independent predictor of unfavorable outcome. Thyroid 2014; 24: 1267–1274.
11. Pon JR, Marra MA. Driver and passenger mutations in cancer. Annu Rev Pathol. 2015; 10: 25–50.
The authors
Laleh Hakima1 DO; Maja H. Oktay1 MD, PhD; Sumanta Goswami*2 PhD
1Department of Cytopathology, Montefiore Medical Center Bronx, NY, USA
2Department of Biology, Yeshiva University, New York, NY, USA
*Corresponding author
E-mail: Goswami@yu.edu
Acute kidney injury is a recognized complication in hospitalized patients and is associated with a high morbidity and high mortality. This brief article aims to summarize the need for early detection of acute kidney injury and the current approach within NHS England to identify such patients.
by Charlotte Fairclough
Background
Acute kidney injury (AKI) is a recognized complication in hospitalized patients. A report in 2009 from National Confidential Enquiry into Patient Outcome and Death (NCEPOD) suggested that AKI was frequently undetected in hospital patients thus contributing to patient morbidity and mortality [1]. Clinical guidelines for recognition and treatment for acute kidney injury were published by NICE (the National Institute for Health and Care Excellence) in 2013 and reported an associated mortality with AKI of more than 25–30% [2]. This guideline also recognized the prevalence of AKI in the primary care population in patients with or without acute illness. NICE also recognized the impact of AKI on healthcare resources, with costs (excluding those in the community) of £434–620 million per year, more than that associated with breast, lung and skin cancer combined [2].
AKI is characterized by an acute loss of the kidney’s excretory capacity leading to accumulation of waste products such as urea and creatinine, and decreased urine output. It is associated with rapid decline in glomerular filtration rate and increases in potassium, phosphate and hydrogen ions. It has varied causes and may be secondary to a non-renal event, thus may be common in hospitalized patients and critically ill patients. It may go undetected in primary care as it can occur without any symptoms. There are associations between co-morbidities, current medications, acute illness and AKI resulting in the high morbidity associated with the condition and the impact on healthcare resources [3].
One of the most common causes of AKI is pre-renal injury due to hypovolemia (a decreased volume of circulating blood). This is thought to be the cause of more than 70% of AKI in the community [4]. This may be exacerbated in patients prescribed certain medications and should be considered carefully by primary care clinicians when assessing patients for AKI [5]. Other causes of AKI are highlighted in Table 1.
Risk factors associated with development of AKI include age, ethnicity, co-morbidities and use of certain medications [3]. It is important to detect the injury as early as possible to prevent the long-term changes in renal function that have been noted to be associated with even less-severe AKI [6].
Defining acute kidney injury
Previous definitions of acute kidney injury had been published, such as RIFLE criteria (Risk Injury, Failure, Loss, End stage renal failure) and AKIN (acute kidney injury network) [7]. KDIGO (Kidney Disease Improving Global Outcomes) published clinical practice guidance in 2011 that categorized AKI based on changes in serum creatinine and/or urine output as defined in both of these previous publications [8]. This categorized AKI into stages 1, 2 and 3 dependent on severity. Evidence suggests that even small, reversible changes in creatinine are associated with worse outcomes, and indeed AKI and severity of AKI is associated with development of chronic kidney disease [6].
The KIDIGO criteria for AKI references changes in creatinine or changes in urine output as a marker for acute kidney injury [8]. Urine output may be the functional marker of kidney function, but can be difficult to monitor. Accurate fluid balance recordings are imperative in management and prevention of AKI in a hospitalized setting, but may be difficult to do accurately especially if the patient is mobile and able to use a toilet unaided. This is also difficult to assess in community patients who obviously will not have recorded urine output as specified in the guidelines. Thus serum creatinine measurements can be used as a marker of kidney function.
Detection of acute kidney injury
Creatinine is used a biomarker for renal function because it is easy and inexpensive to measure. It is also part of most common biochemical panels in blood tests ordered in both hospital and community patients. This means it is easy to monitor trends and to compare to historical data for the patient as required for the diagnosis of AKI. But it may be slow to respond to changes in renal function, and this may be important in the early detection of AKI. Creatinine concentration in the blood and urine is also influenced by other factors such as age, muscle mass, diet, tubular secretion, hydration status and is subject to analytical interferences. Two methods for measuring creatinine are in common use in biochemistry laboratories, the traditional Jaffe methodology and enzymatic methods. Enzymatic methodology for measurement of serum creatinine has been recommended by NICE in the AKI guidelines [2]. As noted above, it has been documented that changes in creatinine only occur when 50% of kidney function has been lost. Therefore, other markers of AKI such as neutrophil gelatinase- associated lipocalin (NGAL) and tissue inhibitors of metalloproteinases- 2 (TIMP-2) have been investigated as alternatives to serum creatinine.
NGAL is a 25-kDa protein in the lipocalin family and is associated with ischaemic kidney injury and may be measured in urine. NGAL is thought to increase in the early stages of AKI as it acts to limit and repair damage caused by the insult and is mediated by NF-κB which is rapidly increased after injury and promotes cell survival and proliferation. It has been found to be detectable in urine in the very early stages of AKI [9].
Tissue inhibitor of metallinoproteinases-2 (TIMP-2) and insulin-like growth factor binding protein 7 (IGFBP7) have been explored as biomarkers of AKI in critically ill patients in an intensive care setting in the Sapphire study [10]. Both of these proteins are inducers of the G1 cell cycle arrest thought to be critical in the development of AKI.
The management of AKI, especially in the community is often focused on removal of the risk factors and inducers of AKI. General Practice can play a role in reduction of the risk of developing AKI such as regular review of those patients on medication associated with increased risk of development of AKI and review of patients with chronic kidney disease who are inherently at increased risk of AKI [5].
NHS England AKI detection algorithm
It was recognized that detecting AKI based on identifying changes in serum creatinine as according to KDIGO guidelines was easily automatable using laboratory information management systems (LIMS). In 2014, NHS England published a patient safety alert to all NHS Trusts with pathology services, to standardize the reporting of AKI [11]. This recognized that some Trusts had already implemented an AKI alert system based on changes in creatinine and the KDIGO guidelines, but aimed to standardize the reporting and ensure reporting was done in real-time.
The alert system algorithm is based on comparison of a patient’s creatinine concentration with that of a baseline creatinine – either a result within the last 48 hours, 7 days or 12 months based on the KDIGO criteria [12]. The patient safety alert algorithm is mandatory for all pathology laboratories in the UK and was developed with the major LIMS providers, thus enabling standardization and a model that is compatible with all systems. The mode of alerting users is not described and thus subject to differing practices within the UK NHS Trusts. This allows for laboratory interaction with users to determine the required practice for each individual Trust. For example the alerts will be reported to the electronic patient record, but whether these results are to be telephoned, emailed, etc., to users is to be individually determined. Implementation into primary care is expected to occur by April 2016 [12].
Conclusion
In summary, AKI is an important issue in healthcare due to the high level of morbidity and mortality associated with it. It is also associated with increased demand on healthcare resources throughout the system including primary and secondary care. Early detection is vital in order to reduce the morbidity and mortality associated with the condition. Every part of the healthcare system, therefore, has a part to play, including GP identification of those patients at increased risk of development of AKI and reduction of that risk, laboratory detection of AKI from serum creatinine measurements or potentially other biomarkers, and to the clinician acting on those alerts and initiating treatment early to preserve renal function.
References
1. Stewart J, Findlay G, Smith N, Kelly K, Mason M. Adding insult to injury. A review of the care of patients who died in hospital with a primary diagnosis of acute kidney injury (acute renal failure). National Confidential Enquiry into Patient Outcome and Death 2009. (http://www.ncepod.org.uk/2009report1/Downloads/AKI_summary.pdf)
2. NICE guidelines CG169. Acute kidney injury: prevention, detection and management. NICE 2013. (https://www.nice.org.uk/guidance/cg169)
3. Wang HE, Muntner P, Chertow GM, Warnock GE. Acute kidney injury and mortality in hospitalized patients. Am J Nephrol. 2012; 35: 349–355.
4. Kaufman J, Dhakal M, Patel B, Hamburger R. Community-acquired acute renal failure. Am J Kidney Dis. 1991; 17(2): 191–198.
5. Blakeman T, Harding S, O’Donoghue D. Acute kidney injury in the community: why primary care has an important role. Br J Gen Pract. 2013; 63(609): 173–174.
6. Chawler LS, Andur R L, Amodeo RL, Kimmel PL, Palant C. The severity of acute kidney injury predicts progression to chronic kidney disease. Kidney Int. 2011; 79 (12): 1361–1369.
7. Lopes JA, Jorge S. The RIFLE and AKIN classifications for acute kidney injury: a critical and comprehensive review. Clin Kidney J. 2013; 6: 8–14.
8. Kidney disease: improving global outcomes (KDIGO) Acute Kidney Injury Work Group. KDIGO Clinical practice guideline for acute kidney injury. Kidney Inter. Suppl. 2012; 2: 1–138.
9. Devarajan P. Neutrophil gelatinase associated lipocalin: a promising biomarker for human acute kidney injury. Biomark Med. 2010; 4(2): 265–280.
10. Pilarczyk K, Edayadiyil-Dudasova M, Wendt D, Demircioglu E, Benedik J, Dohle DS, Jakob H, Duss F. Urinary [TIMP-2]*[IGFBP7] for early prediction of acute kidney injury after coronary artery bypass surgery. Ann intensive care 2015; 5: 50
11. Standardising the early identification of acute kidney injury. NHS England 2014. (https://www.renalreg.org/wp-content/uploads/2014/08/Patient-Safety-Alert-AKI-algorithm-2014_06_04.pdf)
12. -Acute kidney injury warning algorithm best practice guidance. NHS England and UK Renal Registry 2014. (https://www.thinkkidneys.nhs.uk/aki/wp-content/uploads/sites/2/2014/12/AKI-Warning-Algorithm-Best-PracticeGuidance-10.03.16.pdf)
The author
Charlotte Fairclough, MSc
Department of Clinical Chemistry and Metabolic Medicine, Liverpool Clinical Laboratories, Royal Liverpool and Broadgreen University Hospitals NHS Trust,
Liverpool, UK
*Corresponding author
E-mail: charlotte.fairclough@nhs.net
The current prognostic staging of colorectal cancer (CRC) by the tumour, node, metastasis method, alongside the minimal core data set, provides good prognostic information for patient populations but is less accurate for the individual. Reporting of additional histopathological features can improve individualized prognostic staging, but manual microscopic surveillance often results in observer variability and there is a lack of consensus on standardized quantification methods. Automated image analysis can standardize the quantification of prognostic features, can personalize CRC prognosis and augment clinical staging.
by Dr Peter Caie
Introduction
Colorectal cancer (CRC) incidence is extremely high in the developed world, where it is the third most common cancer in men and women in both the UK and USA. There will be an estimated 134 500 new CRC cases diagnosed, and just under 50 000 deaths will have been caused by the disease, in the USA alone. Although the incidence rate has decreased only slightly in the last decade, the change in mortality has dropped significantly between 2000 and 2010, with ~50% of patients across all disease stages surviving for at least 5 years post-diagnosis. Reasons for the decrease in mortality include life style change (47% of CRC cases in the UK could be prevented from healthier lifestyle choice) early detection of disease (e.g. home testing such as fecal occult blood kits), targeted therapy as the result of ‘omics’ research, novel prognostic factors coupled with more accurate pathological and clinical staging of disease and advances in surgical technique. These factors culminate in a more effective treatment of patients at an early stage and at a personalized level. Survival rates in early stage cancer, where the tumour is localized, are extremely good with ~90% of patients experiencing 5-year disease-free survival. Upon spread to localized lymph nodes survival decreases to 50–70% and if distant metastasis has occurred survival is only 12% [1, 2].
Current prognostic staging of CRC
Although multiple CRC subtypes exist, with both molecular and histopathological variances, 90% of CRCs are adenocarcinomas and prognosis is determined through the international tumour, node, metastasis (TNM) staging system alongside the minimal core data set. The TNM staging is based on gross observation and analysis of histopathological tissue sections under the microscope which revolves around the depth of local invasion (T), presence of cancer within the lymph nodes (N) and if the cancer has metastasized (M). TNM staging is excellent at returning prognostic information on a population of patients; however, it is less specialized at predicting prognosis at the level of the individual [3]. A patient’s prognosis is worse the higher the stage they are classified within, however, the TNM system does not differentiate between good and poor outcome of patients within the same stage [4]. There are defined adjuvant treatment guidelines associated with the various stages of CRC [5]. Stage 0 and I cancers will not routinely receive adjuvant chemotherapy and surgical resection is considered curative. Adjuvant therapy is recommended for stage III and IV patients, however, there remains ambiguity about whether to treat all, a subset or no stage II patients with adjuvant chemotherapy [6]. Around 30% of stage II CRC patients will succumb to their disease after surgical resection and, therefore, an accurate and more personalized identification and stratification of high-risk stage II patients, some of whom have comparable or worse outcomes than stage III patients [7], is therefore imperative to increase disease free survival rates. In the UK, pathologists collect a minimal core data set for each patient which helps to identify high-risk stage II CRC cases [8]. Although some parameters within the data set are disregarded in clinical decision making for the management of stage II CRC patients, some features, if present, may invoke the decision to treat the patient: high grade/poor differentiation, pT4 local spread and extramural lymphovascular invasion [9]. However, there is little evidence to date to show the advantage of adjuvant therapy for stage II patients with additional high-risk factors. Furthermore, there are promising histopathological features listed in the literature that have been significantly correlated with poor prognosis but which rarely feature in final clinical reports. There is, for example, growing evidence that immune cell infiltrate and perineural invasion are strongly correlated with poor patient outcome, whereas lymphatic vessel invasion (LVI) and the invasive growth pattern, including tumour budding, are two of the most promising histopathological features that have been significantly associated with lymph node metastasis and disease-specific survival.
Manual reporting yields observer variability
Although histopathological features such as nuclear grade have long been established in the core data sets for CRC prognosis and features such as immune infiltrate, invasive pattern, LVI, lymphatic vessel density (LVD) and the tumour-to-stroma ratio are associated with poor prognosis in the literature, they have also been associated with observer variability in their reporting. Observer variability is an inevitable occurrence when reporting histopathological features by eye under microscopic surveillance; however, it is increased by certain features being obscure under H&E stained tissue with associated retraction artefact, difficult to accurately quantify or if they are rare events. This is particularly true when the calculation of areas in manually determined ‘hot-spots’ is required, such as for LVD and tumour-to-stroma ratio calculations which are both very prone to observer bias and variability. Furthermore, a general consensus on standardized quantification methods is lacking for many of these candidate histopathological features and for this reason they, apart from grading, have not translated into the minimal core data sets of CRC pathological reporting. Although nuclear grading has been reported as a minimal core data set for many years, there have been studies which have also found it to be non-significant [10]. Therefore, there is now a consensus for nuclear grading in CRC to move toward a two-tiered scoring system of ‘well differentiated’ and ‘poorly differentiated’ which eliminates the ‘moderately differentiated’ class and attempts to increase standardization.
Automated image analysis can standardize the quantification of prognostic features
Digital pathology and associated image analysis technology is becoming increasingly sophisticated. It is now possible to create image analysis algorithms that can automatically segment and quantify histopathological features within digital tissue sections with high accuracy. There are multiple advantages to applying image analysis to histopathology reporting that overcome the associated observer variability of manual scoring. Automated image analysis uses standardized algorithms and objectively reports on the features it is programmed to quantify. It does so in a robust manner across all patient samples being tested. Applying image analysis allows a higher degree of accuracy when reporting on the number, area or ratio of specific features across a whole tissue section and so negates the need of manually assigned hot-spots. Furthermore continuous data is captured when image analysis is applied, which allows more accurate clinical cut-offs to be used resulting in a more personalized assessment of a patient’s condition than the more traditional categorical reporting of, e.g. 1+, 2+, 3+ for immunohistochemistry, or ‘well’ or ‘poorly’ differentiated cases. Finally, rare and obscure events that may be missed by the eye are able to be reported with higher confidence when the computer assesses the entire issue section. Jeremy Jass in the late 1980s reported a novel grading system for CRC that included the reporting of the immune infiltrate and the pattern of invasive margin [11]; however, his promising results were not translated into the clinic due to poor reproducibility. Recently, two groups have used image analysis to quantify the immune infiltrate in the form of the immunoscore [12] and the tumour’s infiltrative pattern in the form of tumour budding [13] (Fig. 1A) in a manner that allows standardization, and have shown both features to be prognostically significant. Image analysis has further been used to quantify, amongst others, additional set histopathological features such as for nuclear grading [14], vasculature hot-spots [15] and LVI and LVD (Fig. 1B) [13].
Automated image analysis personalizes CRC prognosis
CRC is a specifically heterogeneous disease and the tumour microenvironment is also a heterogeneous and heterotypic ecology. Therefore, quantifying a single histopathological aspect of a patient’s tumour may not be sufficient for an accurate prognosis across a large population. A recent study by our group used multiplexed immunofluorescence to quantify a suite of histopathological features co-registered across a single CRC tissue section for each patient. Furthermore, the algorithm exported data captured across each nucleus (Fig. 2) in each sample to create a large and complex personalized multi-parametric data set for each patient in the stage II CRC study [16]. A single standardized image analysis algorithm was run across a training set of patients which quantified continuous data from the invasive front of stage II CRC on the number, shape and extent of: tumour buds, poorly differentiated clusters, LVI, LVD, tumour-to-stroma ratio, tumour gland morphology as well as multiple measurements across each nucleus to increase the accuracy of nuclear grading. The resultant ‘big-data’ was distilled through machine learning to identify the optimal parameter set to stratify patients into a high or low risk of disease-specific death. The result was the identification of a novel feature that was independently significant and where the addition of any other measured feature into the model added no further significance to patient stratification. This feature was the mean area of poorly differentiated clusters (area PDC) across the invasive front. The data from the training cohort was validated across a larger independent cohort and again the novel feature held more significance for patient risk stratification in stage II CRC than any of the other more established histopathological feature measured in the study. Furthermore, mathematical modelling was employed to identify if any of the parameters from the clinical pathology report added value to the prediction of disease specific death. By performing this analysis it was found that pT stage and differentiation added further value and were incorporated into a Novel Prognostic Index alongside the area PDC. This novel index outperformed the clinical gold standard of pT staging by almost twofold (Fig. 3).
Big-data and personalized pathology augments clinical staging
The idea behind big-data pathology is to include as much data as possible about each single patient and so to move towards a more personalized prognosis. The acquisition of quantitative data through image analysis and molecular pathology lends itself very well to big-data pathology, where the vast data sets can be mined through sophisticated machine learning algorithms to identify the optimal parameters to answer the clinical question. However, clinical staging and reporting has stood the test of time and it is imperative to include data such as this in any integrative model. As it becomes easier and cheaper to acquire large, reproducible and standardized data sets, modern pathology will become more personalized and patient outcome will improve due to tailored treatment regimens directed at individual patients.
References
1. Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 2011; 61(4): 212–236.
2. Shah R, Jones E, Vidart V, Kuppen PJ, Conti JA, Francis NK. Biomarkers for early detection of colorectal cancer and polyps: systematic review. Cancer Epidemiol Biomarkers Prev. 2014; 23(9):1712–1728.
3. Brenner H, Kloor M, Pox CP. Colorectal cancer. Lancet 2014; 383(9927):1490–1502.
4. Lea D, Haland S, Hagland HR, Soreide K. Accuracy of TNM staging in colorectal cancer: a review of current culprits, the modern role of morphology and stepping-stones for improvements in the molecular era. Scand J Gastroenterol. 2014; 49(10):1153–1163.
5. Poston GJ, Tait D, O’Connell S, Bennett A, Berendse S. Diagnosis and management of colorectal cancer: summary of NICE guidance. BMJ 2011; 343:d6751.
6. Dotan E, Cohen SJ. Challenges in the management of stage II colon cancer. Semin Oncol. 2011; 38(4):511–520.
7. Urquhart R, Bu J, Grunfeld E, Dewar R, MacIntyre M, Porter GA. Examining stage IIB survival in a population-based cohort of patients with colorectal cancer. Cancer 2012; 118(23):5973–5981.
8. Loughrey MB, Quirke P, Shephard NA. Standards and datasets for reporting cancers. Dataset for colorectal cancer histopathology reports July 2014. The Royal College of Pathologists 2014. The cancer datasets are a combination of textual guidance, educational information and reporting proformas to enable consistent grading and staging. (https://www.google.co.uk/?gws_rd=ssl#q=Standards+and+datasets+for+reporting+cancers+Dataset+for+colorectal+cancer+histopathology+reports+July+2014)
9. Morris EJ, Maughan NJ, Forman D, Quirke P. Who to treat with adjuvant therapy in Dukes B/stage II colorectal cancer? The need for high quality pathology. Gut 2007; 56(10):1419–1425.
10. Ratto C, Sofo L, Ippoliti M, Merico M, Doglietto GB, Crucitti F. Prognostic factors in colorectal cancer. Literature review for clinical application. Dis Colon Rectum 1998; 41(8):1033–1049.
11. Jass JR, Love SB, Northover JM. A new prognostic classification of rectal cancer. Lancet 1987; 1(8545):1303–1306.
12. Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, Lugli A, Zlobec I, Hartmann A, et al. Towards the introduction of the Immunoscore in the classification of malignant tumors. J Pathol. 2014; 232(2):199–209.
13. Caie PD, Turnbull AK, Farrington SM, Oniscu A, Harrison DJ. Quantification of tumour budding, lymphatic vessel density and invasion through image analysis in colorectal cancer. J Transl Med. 2014; 12:156.
14. Rathore S, Hussain M, Aksam Iftikhar M, Jalil A. Novel structural descriptors for automated colon cancer detection and grading. Comput Methods Programs Biomed. 2015; 121(2):92–108.
15. Kather JN, Marx A, Reyes-Aldasoro CC, Schad LR, Zollner FG, Weis CA. Continuous representation of tumor microvessel density and detection of angiogenic hotspots in histological whole-slide images. Oncotarget 2015; 6(22):19163–19176.
16. Caie PD, Zhou Y, Turnbull AK, Oniscu A, Harrison DJ. Novel histopathologic feature identified through image analysis augments stage II colorectal cancer clinical reporting. Oncotarget 2016; doi: 10.18632/oncotarget.10053 [Epub ahead of print].
Acknowledgment
This article is based on the author’s recently published paper: Caie PD, Zhou Y, Turnbull AK, Oniscu A, Harrison DJ. Novel histopathologic feature identified through image analysis augments stage II colorectal cancer clinical reporting. Oncotarget 2016; doi: 10.18632/oncotarget.10053 [16].
The author
Peter Caie PhD
Quantitative and Digital Pathology, School of Medicine, University of St Andrews, North Haugh, St Andrews, UK
*Corresponding author
E-mail: Pdc5@st-andrews.ac.uk
Children presenting with diabetic ketoacidosis (DKA) require prompt assessment and treatment initiation to prevent serious complications. The use of point-of-care (POC) analysers to assess blood ketones is beginning to replace the traditional analysis of urine ketones, but some questions remain as to their optimal utilization.
by Dr A.M. Ferguson, Dr J. Michael, Prof. S. DeLurgio and Dr M. Clements
Introduction
Diabetic ketoacidosis (DKA) is an acute complication of uncontrolled diabetes mellitus resulting from insulin deficiency. It is biochemically defined as hyperglycemia (blood glucose >200 mg/dL) with metabolic acidosis (venous pH <7.3 or bicarbonate <15 mmol/L), ketonemia, and ketonuria [1]. The clinical picture of the patient can include fatigue, polydipsia, polyuria, dehydration, abdominal pain, vomiting and altered mental status (Box 1). DKA can occur in known diabetics and can be the presenting symptom prior to diagnosis. Children who are on insulin pump therapy, who have unstable family situations, or have limited access to healthcare are at an increased risk of DKA [1], and DKA is the most common cause of diabetes-related mortality in children.
Assessing urine ketones has been part of the standard practice when assessing if a patient has DKA, but this has multiple issues. There are three types of ketones: acetoacetate, acetone, and β-hydroxybutyrate (BHB). BHB is the predominant ketone produced during DKA and can be present at up to 10 times the amount of acetoacetate. The urine dipsticks that are commonly used to assess ketonuria utilize a nitroprusside reagent that reacts with acetoacetate and acetone but not at all with BHB. This is problematic because the major ketone produced in DKA is not detected, which can lead to false negative urine ketone testing. Additionally, as ketosis resolves, BHB is converted to acetoacetate, increasing urine ketones during the recovery phase, potentially leading the clinician to believe that the ketosis is worsening instead of resolving. An added obstacle is the difficulty of getting a urine specimen from a young child, especially one in nappies. Measuring serum ketones, specifically BHB, is a solution to both of these issues.
Clinical measurement of serum ketones
As the methodology for measuring serum BHB became more automated, the test moved from being used only on a research basis to being available for clinical use. Initial studies were done to see how serum BHB functioned for the diagnosis of DKA. A large retrospective study looking at simultaneous measurements of BHB and bicarbonate found that BHB levels of ≥3 and ≥3.8 mmol/L in children and adults, respectively, could be used to diagnose DKA and provides a more specific assessment of DKA than bicarbonate alone [2].
When assessing patients for DKA, it is critical to make the diagnosis as quickly as possible to initiate treatment and prevent the patient from decompensating further. The commercial availability of point-of-care (POC) meters to assess serum ketones allows the patient to be tested immediately on presentation at the bedside. There have been multiple studies performed in adults showing that use of POC BHB meters in the emergency room can aid in diagnosis and treatment of DKA. Arora et al. compared POC BHB and urine ketone dipstick results in 54 patients with DKA presenting to the emergency department [3]. They found that both methods were equally sensitive for detecting DKA at 98.1%, but that BHB with a cut-off of ≥1.5 mmol/L is more specific for DKA compared to urine dipsticks (78.6 vs 35.1%) and could cut down on unnecessary DKA work ups in hyperglycemic patients. Another study found that a BHB value of 3.5 mmol/L yielded 100% sensitivity and specificity for the diagnosis of DKA [4].
Use of POC testing in pediatrics
Fewer studies have been done in pediatric patients. One such study by Ham et al. determined that using a POC meter in the hospital setting could aid in monitoring the resolution of DKA in pediatric patients [5]. The BHB values from the POC meter correlated with BHB values from the laboratory for most of the meter’s measurement range. Use of the meter had both a strong positive predictive value (PPV, 0.85) as well as negative predictive value (NPV, 1.0) for indicating the presence or absence of DKA at a meter value of 1.5 mmol/L [5]. Noyes et al. used POC ketone testing to identify the endpoint of an integrated care pathway when treating DKA in children [6]. They compared their current treatment endpoint of pH >7.3 and no presence of urine ketones with an endpoint defined by pH >7.3 and two successive POC ketone measurements of <1 mmol/L. The study measured time of treatment in 35 patient episodes in children ranging in age from 1–14 years. The time to completion of treatment using POC ketone measurement was 17 hours, compared to 28 hours using measurement of urine ketones to end treatment [6] . They found that occasionally a value below 1 mmol/L would be followed by a value above 1 mmol/L, but this never occurred after two subsequent values under 1 mmol/L, leading them to recommend waiting for the two successive low values before ending treatment. In addition to allowing an earlier treatment endpoint, this approach enables less time to be spent in the ICU, with decreased cost associated with treatment. Using a POC ketone meter can also result in fewer tests being ordered overall. Rewers and colleagues asked whether monitoring serum BHB values at the bedside could result in a decrease in laboratory testing in pediatric patients [7]. Their results indicated that the real-time changes observed in POC serum BHB values correlated strongly with changes in pH, bicarbonate, and pCO2 and also had good correlation with the laboratory BHB method. While initial measurement of pH, bicarbonate and pCO2 is encouraged, following up the patient with POC BHB can replace serial laboratory measurements of those analytes and decrease the amount of laboratory testing [7]. Similarly, a separate study showed that use of a POC BHB meter at home decreased diabetes-related hospital visits and hospitalizations of pediatric diabetics when compared to urine ketone testing by allowing earlier identification of ketosis and initiation of treatment [8].
Most of the studies mentioned are close to 10 years old, but measuring serum BHB to diagnose DKA or monitor its resolution has not become standard practice. A recent review of the standard treatment guidelines for DKA in children and adolescents raises the question of whether blood ketones should be evaluated during management of DKA [9]. The authors recommend using serum BHB measurement, either from the laboratory or at the point of care, to both diagnose DKA and monitor treatment. Despite the inaccuracies of POC meters seen at high BHB values [5–7], use of a diagnostic cut-off of >3 mmol/L is well within the accurate range of the meters and can be used to confidently diagnose DKA and monitor the patient’s response to treatment.
Conclusions
Despite the increasing body of knowledge indicating that measurement of serum BHB can aid in both diagnosis and management of DKA, a study conducted in 2014 indicated that although 89% of pediatric emergency medicine and critical care providers responding to a survey stated that they had a DKA protocol at their institution, 67% perceived no clinical advantage in the use of serum ketone measurements [10]. This suggests that evaluation of serum ketone monitoring during DKA management from a quality improvement and research perspective may be necessary before clinical adoption is widespread. The next iteration of DKA management guidelines should address the potential utility of serum ketone monitoring.
References
1. Wolfsdorf J, Craig ME, et al. Diabetic ketoacidosis in children and adolescents with diabetes. Pediatr Diabetes 2009; 10(Suppl 12): 118–133.
2. Sheikh-Ali M, Karon BS, et al. Can serum beta-hydroxybutyrate be used to diagnose diabetic ketoacidosis? Diabetes Care 2008; 31(4): 643–647.
3. Arora S, Henderson SO, et al. Diagnostic accuracy of point-of-care testing for diabetic ketoacidosis at emergency-department triage: {beta}-hydroxybutyrate versus the urine dipstick. Diabetes Care 2011; 34(4): 852–854.
4. Charles RA, Bee YM, et al. Point-of-care blood ketone testing: screening for diabetic ketoacidosis at the emergency department. Singapore Med J. 2007; 48(11): 986–989.
5. Ham MR, Okada P, White PC. Bedside ketone determination in diabetic children with hyperglycemia and ketosis in the acute care setting. Pediatr Diabetes 2004; 5(1): 39–43.
6. Noyes KJ, Crofton P, et al. Hydroxybutyrate near-patient testing to evaluate a new end-point for intravenous insulin therapy in the treatment of diabetic ketoacidosis in children. Pediatr Diabetes 2007; 8(3): 150–156.
7. Rewers A, McFann K, Chase HP. Bedside monitoring of blood beta-hydroxybutyrate levels in the management of diabetic ketoacidosis in children. Diabetes Technology & Therapeutics 2006; 8(6): 671–676.
8. Laffel LM, Wentzell K, et al. Sick day management using blood 3-hydroxybutyrate (3-OHB) compared with urine ketone monitoring reduces hospital visits in young people with T1DM: a randomized clinical trial. Diabet Med. 2006; 23(3): 278–284.
9. Wolfsdorf JI. The International Society of Pediatric and Adolescent Diabetes guidelines for management of diabetic ketoacidosis: Do the guidelines need to be modified? Pediatr Diabetes 2014; 15(4): 277–286.
10. Clark MG, Dalabih A. Variability of DKA management among pediatric emergency room and critical care providers: a call for more evidence-based and cost-effective care? J Clin Res Pediatr Endocrinol. 2014; 6(3): 190–191.
The authors
Angela M. Ferguson*1 PhD, DABCC, FACB; Jeffery Michael1 D.O., FAAP; Stephen DeLurgio2 PhD; Mark Clements1 MD, PhD, CPI
1Children’s Mercy Hospital, Kansas City, MO, USA
2Bloch School, University of Missouri, Kansas City, MO, USA
*Corresponding author
E-mail: amferguson@cmh.edu
A diagnostic point-of care (POC) test would greatly improve case detection and management of pediatric tuberculosis (TB). Herein, we provide a brief overview of the challenges associated with diagnosing TB in children and recent evidence that points towards a future POC test based on transcriptomes in peripheral blood.
by Dr S. Jenum, Dr J.E. Gjøen, R. Bakken, Dr D. Sivakumaran and Prof. H.M.S. Grewal
Introduction
Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb), is a leading cause of childhood death and morbidity worldwide estimated to cause 1 million new cases in children <15 years of age in 2014 [1]. The gold standard for a diagnosis of pulmonary TB is growth of Mtb from respiratory specimens. However, culture-confirmed TB only occurs in about 30% of pediatric TB cases because of the paucibacillary nature of the disease and the difficulties in obtaining representative specimens [2]. For these reasons, children have been perceived less contagious and, therefore, not considered a priority for case finding within national TB programmes. This has resulted in detection rates being as low as 35% [3], with evident consequences for childhood death and morbidity. An accurate diagnostic test that provides a rapid, sensitive and specific result, enabling early initiation of treatment at the point-of-care (POC), ‘a diagnostic POC test’, could prevent this. The declaration by the World Health Organization (WHO) of pediatric TB as a neglected area of science and the stated urgent need for improved diagnostics [4], has spurred the search for TB diagnostic biomarkers in children. We provide a brief overview of the challenges associated with diagnosing TB in children and recent evidence that points towards a future POC test based on transcriptomes in peripheral blood.
Diagnosing TB in children
Diagnostic approaches and criteria for pediatric TB have recently been extensively reviewed [5]. In summary, sampling of respiratory specimens for confirmation of diagnosis by culture is strongly recommended, but requires repeated sampling of induced sputum and gastric aspirates, which are cost and labour intensive [5]. Notably, as many as 70% of children are culture negative even under optimized study conditions [2]. Therefore, in most cases, a diagnosis is based on a diagnostic algorithm including clinical signs and symptoms, radiological findings suggestive of TB and immunological evidence of previous Mtb sensitization [5]. Clinical diagnostic algorithms have not being adequately validated [6], and radiological findings are often unspecific [5]. The tuberculin skin test (TST) and the newer interferon-γ release assays used as a surrogate for Mtb infection cannot differentiate between infection and disease [7], and have reduced sensitivity in young and malnourished children [8, 9], but may be complementary by improving sensitivity in such clinical contexts [5].
Requirements for a POC test
Direct microscopy of sputum smears looking for acid-fast bacilli, is widely implemented at primary diagnostic centres for the diagnosis of pulmonary TB in adults, but the sensitivity is highly variable (20–80%) [10]. Since 2010, the use of the Xpert MTB/RIF assay on sputum samples has expanded in low- and middle-income countries [11], serving as a POC test in adults. The assay is based on direct identification of Mtb in specimens by a nucleic acid amplification test and detects in children three times the cases detected by direct microscopy and ~70% of the cases detected by liquid culture [12]. The WHO recommends that a POC triage test, meant to narrow down the population that needs further diagnostic work-up, should achieve a sensitivity of 90% and a specificity of 70%. For a diagnostic POC test, the sensitivity should be >95% and the specificity >98% [13]. A POC test based on Mtb detection will never meet these requirements in children because of the high proportion of culture-negative pulmonary TB [2], as well as the significant proportion of extrapulmonary TB [5]. Host biomarkers reflecting the ongoing pathological processes resulting from Mtb infection may hold greater promise for this purpose. Moreover, peripheral whole blood (WB) is more easily available for diagnosis than respiratory or tissue samples, particularly in children. Therefore, biomarkers based on transcriptomes in WB have gained increasing interest.
Genome-wide analysis of RNA expression
A landmark study by Berry et al., based on genome-wide analysis of RNA expression in unstimulated WB from adults, identified an 86-transcript signature able to discriminate active TB from other inflammatory and infectious diseases [14]. This transcriptomic signature consisted mainly of neutrophil-driven interferon (IFN)-inducible genes, which until then, had not been considered essential players in TB immunity. Further, a decreased abundance of B-cell transcripts in TB patients [14], challenged the paradigm of B-cells and humoral immunity being of little importance in protection against Mtb. Following this study, the upregulation of type I IFN-inducible genes and altered expression of B-cell related genes has been confirmed in adults [15–19] and adolescents [20].
To our knowledge, two previous studies in children have been published based on genome-wide transcriptional profiling of WB in analysis of RNA expression in Warao Amerindians [21], and in three African cohorts [22]. Verhagen et al. addressed the diagnostic gap in discriminating TB disease from latent TB, and identified a 5-transcript signature with a prediction error of 11% in discriminating active from latent TB. The signature classified correctly 78% of TB cases and 100% of latent TB cases in a validation cohort comprising children with non-TB pneumonia [21]. Interestingly, the 5-transcript signature, when applied on transcriptional data-sets generated by microarray analyses of WB from adults performed similarly, whereas the gene signatures identified in these adults were useless in the cohort of Warao Amerindian children [21]. This questions the validity in extrapolating results from transcriptional TB biomarker research in adults to children, highlighting the importance of studies in pediatric populations.
Applying a more real-life diagnostic setting with inclusion of children evaluated for suspected TB applying a graded diagnosis of TB in line with the consensus statement [6], Anderson et al. identified a 51-transcript signature that distinguished TB from other diseases in South African and Malawian children, subsequently validated in Kenyan children. A risk score based on the signature identified confirmed TB with a sensitivity of 82.9% and a specificity of 83.6%, and discriminated between other disease and culture-negative TB with a sensitivity ranging from 35–82% [22].
A more cost-effective method for assessing transcriptomes
Genome-wide analyses of transcriptomes are costly and resource-intensive but can be seen as a necessary first step in identifying markers with potential for subsequent refinement as POC tests. A test more suitable for hypothesis-testing or more high-throughput screening of transcriptional signatures with relevance in TB, has recently been developed [23]. The dual-colour Reverse Transcriptase-Multiplex Ligation-dependent Probe Amplification (dcRT-MLPA) can rapidly profile multiple host genes with a dynamic range and accuracy comparable to real-time qPCR and RNA sequencing at a cost of €5–7 per sample [24], most likely simplifying the translation of findings into diagnostic tools in resource-poor settings. The dcRT-MLPA method has been applied in different studies assessing TB biomarkers [19, 23–25]. Recently, incorporating the results from unbiased gene-expression profiling, the gene panels adapted for the dcRT-MLPA platform have been expanded to include type I IFN-inducible genes as well as genes covering adaptive immunity, including B-cells [25].
The dcRT-MLPA method in the context of pediatric TB
We have applied the dcRT-MLPA method to identify TB diagnostic transcriptome signatures in pediatric TB settings in India [26, 27]. We have also examined the immunological basis for discordant TST and Quantiferon® responses [28], as well as the potential interference of non-tuberculous mycobacteria on transcriptional biomarkers [29]. The younger the child, the more challenging the diagnosis of TB. We therefore explored transcriptional biomarkers in Indian children aged <3 years referred for a TB diagnostic work-up in a prospective cohort study of BCG-vaccinated neonates. We found that RAB33A alone discriminated between clinical TB and Mtb infection with an area under the curve (AUC) of 77.5%, whereas a 5-transcript signature effectively discriminated between clinical TB and controls (AUC 91.7%) [26]. Then, in a cohort of older Indian children (mean age ~9 years) with intrathoracic TB and their siblings, we assessed transcriptional biomarkers across the spectrum of TB disease. We identified 12 biomarkers consistently associated with clinical groups ‘upstream’ towards confirmed TB or ‘downstream’ towards a decreased likelihood of TB disease (Fig. 1), suggesting a correlation with Mtb-related pathology and high relevance to a future POC test. Furthermore, an 8-transcript signature separated children with TB from asymptomatic siblings (AUC 88%) [27]. Notably, these transcriptome signatures provided a superior sensitivity for confirmed TB compared with the Xpert MTB/RIF performed on gastric lavage [2]. Many of the biomarkers within these signatures have been confirmed in adults with Mtb infection or disease [19, 23, 24].
Future perspectives
We are currently exploring the performance of the extended dcRT-MLPA panels of genes which include type I IFN-signalling, myeloid cell activation, general inflammation and B-cell related genes in Indian children with intrathoracic TB. Transcriptome signatures identified in a discovery set consisting of randomly assigned TB cases and their healthy siblings will be validated in a diagnostic setting of symptomatic children aged <3 years.
We are also exploring the association between transcriptional biomarkers and the extent of TB disease and the outcome of TB treatment in children. The monitoring and evaluation of TB treatment is hampered by the limited tools available to guide decision-making during the long treatment period. Based on our unpublished results, as well as from findings in adults [16, 17, 23] we believe that treatment monitoring by means other than a negative culture result at 2 months post-treatment [30] will be possible.
Conclusions
Transcriptome-based tests meeting the requirements for both a screening and a diagnostic POC test would greatly improve case detection in children and subsequently reduce morbidity and mortality attributable to TB [13]. Further, a POC test capable of guiding treatment would prevent treatment failure and the subsequent emergence of resistant Mtb strains.
References
1. WHO. Global tuberculosis report 2015. ISBN 978-9241565059. http://apps.who.int/iris/bitstream/10665/191102/1/9789241565059_eng.pdf.
2. Dodd LE, Wilkinson RJ. Diagnosis of paediatric tuberculosis: the culture conundrum. Lancet Infect Dis. 2013; 13(1): 3–4.
3. Dodd PJ, Gardiner E, Coghlan R, Seddon JA. Burden of childhood tuberculosis in 22 high-burden countries: a mathematical modelling study. Lancet Glob Health 2014; 2(8): e453–459.
4. WHO. Roadmap for childhood tube culosis: towards zero deaths. WHO 2013;http://apps.who.int/iris bitst eam/10665/89506/1/9789241506137_eng pdf.
5. Perez-Velez CM, Marais BJ. Tuberculosis in children. N Engl J Med. 2012; 367(4): 348–361.
6. Graham SM, Ahmed T, Amanullah F, Browning R, Cardenas V, Casenghi M, Cuevas LE, Gale M, Gie RP, et al. Evaluation of tuberculosis diagnostics in children: 1. Proposed clinical case definitions for classification of intrathoracic tuberculosis disease. Consensus from an expert panel. J Infect Dis. 2012; 205 Suppl 2: S199–S208.
7. Pai M, Denkinger CM, Kik SV, Rangaka MX, Zwerling A, Oxlade O, Metcalfe JZ, Cattamanchi A, Dowdy DW, et al. Gamma interferon release assays for detection of Mycobacterium tuberculosis infection. Clin Microbiol Rev. 2014; 27(1): 3–20.
8. Machingaidze S, Wiysonge CS, Gonzalez-Angulo Y, Hatherill M, Moyo S, Hanekom W, Mahomed H. The utility of an interferon gamma release assay for diagnosis of latent tuberculosis infection and disease in children: a systematic review and meta-analysis. Pediatr Infect Dis J. 2011; 30(8): 694–700.
9. Jenum S, Selvam S, Mahelai D, Jesuraj N, Cardenas V, Kenneth J, Hesseling AC, Doherty TM, Vaz M, Grewal HM. Influence of age and nutritional status on the performance of the tuberculin skin test and QuantiFERON-TB gold in-tube in young children evaluated for tuberculosis in Southern India. Pediatr Infect Dis J. 2014; 33(10): e260–269.
10. WHO. Report of the Tenth Meeting of the Strategic and Technical Advisory Group for Tuberculosis (STAG-TB) 2010. WHO 2010; http://www.who.int/tb/advisory_bodies/stag_tb_report_2010.pdf.
11. Qin ZZ, Pai M, Van GW, Sahu S, Ghiasi M, Creswell J. How is Xpert MTB/RIF being implemented in 22 high tuberculosis burden countries? Eur Respir J. 2015; 45(2): 549–554.
12. Nicol MP, Workman L, Isaacs W, Munro J, Black F, Eley B, Boehme CC, Zemanay W, Zar HJ. Accuracy of the Xpert MTB/RIF test for the diagnosis of pulmonary tuberculosis in children admitted to hospital in Cape Town, South Africa: a descriptive study. Lancet Infect Dis. 2011; 11(11): 819–824.
13. WHO. Global Tuberculosis Report. WHO 2014; http://apps.who.int/iris/bitst eam/10665/137094/1/9789241564809_eng.pdf?ua=1.
14. Berry MP, Graham CM, McNab FW, Xu Z, Bloch SA, Oni T, Wilkinson KA, Banchereau R, Skinner J, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature 2010; 466(7309): 973–977.
15. Ottenhoff TH, Dass RH, Yang N, Zhang MM, Wong HE, Sahiratmadja E, Khor CC, Alisjahbana B, van Crevel R, et al. Genome-wide expression profiling identifies type 1 interferon response pathways in active tuberculosis. PLoS One 2012; 7(9): e45839.
16. Bloom CI, Graham CM, Berry MP, Wilkinson KA, Oni T, Rozakeas F, Xu Z, Rossello-Urgell J, Chaussabel D, et al. Detectable changes in the blood transcriptome are present after two weeks of antituberculosis therapy. PLoS One 2012; 7(10): e46191.
17. Cliff JM, Lee JS, Constantinou N, Cho JE, Clark TG, Ronacher K, King EC, Lukey PT, Duncan K, et al. Distinct phases of blood gene expression pattern through tuberculosis treatment reflect modulation of the humoral immune response. J Infect Dis. 2013; 207(1): 18–29.
18. Kaforou M, Wright VJ, Oni T, French N, Anderson ST, Bangani N, Banwell CM, Brent AJ, Crampin AC, et al. Detection of tuberculosis in HIV-infected and -uninfected African adults using whole blood RNA expression signatures: a case-control study. PLoS Med. 2013; 10(10): e1001538.
19. Sutherland JS, Loxton AG, Haks MC, Kassa D, Ambrose L, Lee JS, Ran L, van Baarle D, Maertzdorf J, et al. Differential gene expression of activating Fcγ receptor classifies active tuberculosis regardless of human immunodeficiency virus status or ethnicity. Clin Microbiol Infect. 2014; 20(4): O230–238.
20. Zak DE, Penn-Nicholson A, Scriba TJ, Thompson E, Suliman S, Amon LM, Mahomed H, Erasmus M, Whatney W, et al. A blood RNA signature for tuberculosis disease risk: a prospective cohort study. Lancet 2016; 387(10035): 2312–2322.
21. Verhagen LM, Zomer A, Maes M, Villalba JA, Del Nogal B, Eleveld M, van Hijum SA, de Waard JH, Hermans PW. A predictive signature gene set for discriminating active from latent tuberculosis in Warao Amerindian children. BMC Genomics 2013; 14: 74.
22. Anderson ST, Kaforou M, Brent AJ, Wright VJ, Banwell CM, Chagaluka G, Crampin AC, Dockrell HM, French N, et al. Diagnosis of childhood tuberculosis and host RNA expression in Africa. New Eng J Med. 2014; 370(18): 1712–1723.
23. Joosten SA, Goeman JJ, Sutherland JS, Opmeer L, de Boer KG, Jacobsen M, Kaufmann SH, Finos L, Magis-Escurra C, et al. Identification of biomarkers for tuberculosis disease using a novel dual-color RT-MLPA assay. Genes Immun. 2012; 13(1): 71–82.
24. Haks MC, Goeman JJ, Magis-Escurra C, Ottenhoff TH. Focused human gene expression profiling using dual-color reverse transcriptase multiplex ligation-dependent probe amplification. Vaccine 2015; 33(40): 5282–5288.
25. Sloot R, Schim van der Loeff MF, van Zwet EW, Haks MC, Keizer ST, Scholing M, Ottenhoff TH, Borgdorff MW, Joosten SA. Biomarkers can identify pulmonary tuberculosis in HIV-infected drug users months prior to clinical diagnosis. EBioMedicine 2015; 2(2): 172–179.
26. Dhanasekaran S, Jenum S, Stavrum R, Ritz C, Faurholt-Jepsen D, Kenneth J, Vaz M, Grewal HM, Doherty TM; TB Trials Study Group. Identification of biomarkers for Mycobacterium tuberculosis infection and disease in BCG-vaccinated young children in Southern India. Genes Immun. 2013; 14(6): 356–364.
27. Jenum S, Dhanasekaran S, Lodha R, Mukherjee A, Kumar Saini D, Singh S, Singh V, Medigeshi G, Haks MC, et al. Approaching a diagnostic point-of-care test for pediatric tuberculosis through evaluation of immune biomarkers across the clinical disease spectrum. Sci Rep. 2016; 6: 18520.
28. Dhanasekaran S, Jenum S, Stavrum R, Ritz C, Kenneth J, Vaz M, Doherty TM, Grewal HM; TB Trials Study Group. Concordant or discordant results by the tuberculin skin test and the quantiFERON-TB test in children reflect immune biomarker profiles. Genes Immun. 2014; 15(5): 265–274.
29. Dhanasekaran S, Jenum S, Stavrum R, Wiker HG, Kenneth J, Vaz M, Doherty TM, Grewal HM; TB Trials Study Group. Effect of non-tuberculous Mycobacteria on host biomarkers potentially relevant for tuberculosis management. PLoS Negl Trop Dis. 2014; 8(10): e3243.
30. Wallis RS, Kim P, Cole S, Hanna D, Andrade BB, Maeurer M, et al. Tuberculosis biomarkers discovery: developments, needs, and challenges. Lancet Infect Dis. 2013; 13(4): 362–372.
The authors
Synne Jenum*1 MD, PhD; John Espen Gjøen2 MD, Rasmus Bakken2; Dhanasekaran Sivakumaran2 PhD; Harleen M.S. Grewal2 MD, PhD, DTMH
1Department of Infectious Diseases, Oslo University Hospital, Pb 4950, N-0424 Oslo, Norway
2Department of Clinical Science, University of Bergen, Pb 7804, N-5021 Bergen, Norway
*Corresponding author
E-mail: synnejenum@gmail.com
The aim of this study was to perform proteomic analysis of serum from pediatric patients with B-cell acute lymphoblastic leukemia (B-ALL) to identify candidate biomarker proteins, for use in early diagnosis and evaluation of treatment. This approach is an alternative to traditional techniques that can investigate the disease from another perspective. Acute lymphoblastic leukemia is the most common malignant cancer in childhood and the symptoms of childhood cancer are difficult to recognize.
by Dr M. de S. Cavalcante, Prof. A. E. Vieira-Neto,
Dr R. de A. Moreira and Dr A. C. de O. Monteiro-Moreira
Background and significance
Acute lymphoblastic leukemia (ALL) is the most common malignant cancer in childhood, and is responsible for approximately 25% of all childhood cancers and 72% of all cases of pediatric leukemia [1]. The current standards for diagnosis of ALL integrate the study of cell morphology, immunophenotyping and genetics/cytogenetics, as described in the classification of lymphoid cancers published by the World Health Organization (WHO) in 2008 [2]. Of lymphoid cancers, as designated using the most recent WHO classification, the purely leukemic presentation, B-lineage ALL (85 %) is the most common [3], and will be addressed in this study. The signs and symptoms of childhood cancer are very challenging to identify, as it is not the first diagnosis to be considered for nonspecific complaints, leading to potential uncertainty in diagnosis. Moreover, children showing the first signs of cancer frequently do not appear severely ill, which may delay diagnosis. In addition, childhood cancer can mimic other common childhood diseases and even normal developmental physiological processes [4]. In the specific case of ALL, early diagnosis and treatment increase the chances of a cure [4].
Future prospects
A label-free proteomic approach was used for the quantitative analysis. Other approaches could also be used in the future, for example it is possible to find studies using RNA interference, mainly silencing expression of specific genes [5]. In our proteomic approach, for each protein, the program ExpressionE selected all corresponding peptides from the samples and compared the intensities of these for relative protein quantification. Using the intensity of a peptide of known quantity, alcohol dehydrogenase (ADH), the program performed self-standardization of data sets. Lists of proteins were then filtered to show only those present in all three repeated injections of each sample, from which an output table was created. This table showed the names, access codes, and expression levels of the proteins, and indicated whether they were upregulated ≥2-fold, downregulated ≤0.5-fold, or whether they did not show significant differences between the groups (unchanged), 0.5 < expression level < 2. The list of proteins generated from three injections of samples in MS, coupled with broad limits used for protein expression levels and serum samples used the controls (non-leukemic pediatric patients) may suggest that the panel of candidate protein biomarkers is clearly increased in the disease state.
Biotechnological resources
Affinity chromatography with α-D-galactose-binding lectin from Artocarpus incisa [6] immobilized on a SepharoseTM 4B gel, combined with identification and quantification of glycoproteins by mass spectrometry, are excellent tools for comparative serum studies. The biomarker pipeline is commonly viewed as a series of preclinical phases: biomarker discovery, and verification before the final clinical evaluation. The comparative analysis results in a list of hundreds of proteins that are differentially expressed between healthy and diseased samples [7]. In this study, the preclinical phase of biomarker discovery was applied and a proteomic analysis of serum samples from pediatric patients with B-ALL was performed, to analyse levels of glycoprotein expression, with the aim of identifying biomarkers to aid in the early diagnosis of B-ALL and to assess the response to induction therapy.
The depletion of high-abundance proteins in serum, human serum albumin (HSA) and IgG, followed by affinity chromatography with the plant lectin Frutalin immobilized on SepharoseTM 4B (Fig. 1), reduced the dynamic range and increased the capacity to identify lower-abundance proteins. The retained fraction (FR) peak containing the protein of interest was concentrated and digested, for later analysis by nano-LC-MS/MS.
Proteomic approach
The study population was composed mainly of children from the lower middle class, who attended a reference hospital for the diagnosis and treatment of childhood cancers in the State of Ceará, Brazil. The study was conducted with the approval of the Research Ethics Committee at the Hospital Infantil Albert Sabin, associated with the Secretary of Health of the State of Ceará. The demographic and clinical data for the patients are summarized in Table 1. The pediatric patients were evaluated at two different times: at diagnosis (B-ALL group; n = 10) and after induction therapy (AIT group; n = 10). Samples of healthy children (Control group; n = 10) were obtained for comparison.
The differentially expressed proteins were used for pathway analysis. Swiss-Prot accession numbers were inserted into the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) software, version 9.05 (available at http://string.embl.de/), with the following analysis parameters: Homo sapiens, confidence level 0.400–0.900, using the active prediction method [8].
Biomarker panel for ALL diagnosis
A panel of protein biomarker candidates has been developed for pre-diagnosis of B-ALL and also provide information that would indicate a favourable response to treatment after induction therapy. In the proteomic analysis, a total of 96 proteins were identified. Leucine-rich alpha-2-glycoprotein 1 (LRG1), Clusterin (CLU), thrombin (F2), heparin cofactor II (SERPIND1), alpha-2-macroglobulin (A2M), alpha-2-antiplasmin (SERPINF2), Alpha-1 antitrypsin (SERPINA1), Complement factor B (CFB) and Complement C3 (C3) were over-expressed in the B-ALL compared to the Control and AIT groups, and were, therefore, identified as candidate biomarkers for early diagnosis of B-ALL. The AIT group showed no significant differences in the expression levels of these proteins compared to the Control group, and did not show any significant change in the level of expression of these proteins, a fact that further reaffirms the presence of these potential biomarkers in a disease state, as all patients achieved complete remission after treatment (Fig. 2). Our results also confirm the important relationship between cancer and phenomena associated with blood coagulation. Several studies have reported that approximately 50% of patients with malignant disease and more than 90% of those that evolve to metastasis present evidence of abnormalities in coagulation and/or fibrinolysis [9–13].
Conclusion
Acute lymphoblastic leukemia is the most common malignant cancer in childhood and this proteomic approach is an alternative to traditional techniques, since the signs and symptoms of childhood cancer are very challenging to identify. LRG1, CLU, F2, SERPIND1, A2M, SERPINF2, SERPINA1, CFB, and C3 were identified as candidate biomarkers for early diagnosis of B-ALL; all were over-expressed in the B-ALL group compared to the Control and AIT groups. The AIT group did not display any significant changes in the expression levels of these proteins, compared to the Control group. All patients in the AIT group achieved complete remission after treatment; this indicates that these biomarkers are only present in the disease state. These candidate biomarkers may improve the pre-diagnosis of B-ALL, which is currently difficult to diagnose in the early stages; the biomarkers may also provide key information on the response to treatment after induction therapy. Further clinical and genomic studies will be important to improve the survival of children with this disease.
Acknowledgements
FINEP, CNPq, RENORBIO-UNIFOR, ALBERT SABIN HOSPITAL
This article is a summary of a paper first published in Biomarker Research: Cavalcante Mde S, Torres-Romero JC, Lobo MD, Moreno FB, Bezerra LP, Lima DS, Matos JC, Moreira Rde A, Monteiro-Moreira AC. A panel of glycoproteins as candidate biomarkers for early diagnosis and treatment evaluation of B-cell acute lymphoblastic leukemia. Biomarker Research 2016; 4: 1 (doi: 10.1186/s40364-016-0055-6) [14].
References
1. Scheurer ME, Bondy ML, Gurney JG. Epidemiology of Childhood Cancer. In: Pizzo PA, Poplack DG, editors. Principles and practice of pediatric oncology, 6th ed, pp2–16. Lippincott Williams and Wilkins 2011.
2. Vardiman JW, Thiele J, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 2009; 114: 937–951.
3. Chiaretti S, Zini G, Bassan R. Diagnosis and subclassification of acute lymphoblastic leukemia. Mediterr J Hematol Infect Dis. 2014; 6: e2014073.
4. Rodrigues KE, Camargo B. Diagnóstico precoce do câncer infantil: responsabilidade de todos. Rev Assoc Med Bras. 2003; 49: 29–34 (in Portuguese).
5. Trougakos IP, So A, et al. Silencing expression of the clusterin/apolipoprotein j gene in human cancer cells using small interfering RNA induces spontaneous apoptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res. 2004; 64: 1834–1842.
6. Monteiro-Moreira ACO, Pereira HD, et al. Crystallization and preliminary x-ray diffraction studies of Frutalin, an α-D-galactose-binding lectin from Artocarpus incisa seeds. Acta Crystallographica Session F, 2015.
7. Parker CE, Borchers CH. Mass spectrometry based biomarker discovery, verification, and validation–quality assurance and control of protein biomarker assays. Mol Oncol. 2014; 8(4): 840–858.
8. Jensen LJ, Kuhn M, et al. STRING 8–a global view on proteins and their functional interactions in 630 organisms. Nucleic Acids Res. 2009; 37: D412–416.
9. Kwon H-C, Oh SY, et al. Plasma levels of prothrombin fragment F112, D-dimer and prothrombin time correlate with clinical stage and lymph node metastasis in operable gastric cancer patients. Jpn J Clin Oncol. 2008; 38: 2–7.
10. Bick RL. Coagulation abnormalities in malignancy: a review. Semin Thromb Hemost. 1992; 18: 353–372.
11. Luzzatto G, Schafer Al. The prethrombotic state in cancer. Semin Oncol. 1990; 17: 147–159.
12. Nigel O’Connor, Gozzard DI, et al. Haemostatic abnormalities and malignant disease. Lancet 1986; 8: 303–304.
13. Hillen HF. Thrombosis in cancer patients. Ann Oncol. 2000; 11: 273–276.
14. Cavalcante Mde S, Torres-Romero JC, et al. A panel of glycoproteins as candidate biomarkers for early diagnosis and treatment evaluation of B-cell acute lymphoblastic leukemia. Biomarker Research 2016; 4: 1 (doi: 10.1186/s40364-016-0055-6).
The authors
Márcio de Souza Cavalcante1, Antonio Eufrásio Vieira-Neto², Renato de Azevedo Moreira3, Ana Cristina de Oliveira Monteiro-Moreira3*
1Northeast Network of Biotechnology (RENORBIO), State University of Ceará, Ceará, Brazil.
2Center of Experimental Biology (NUBEX), University of Fortaleza (UNIFOR), Ceará, Brazil.
3Department of Biochemistry and Molecular Biology, Federal University of Ceará, Ceará, Brazil.
4Development and Technological Innovation in Drug Program, Federal University of Ceará, Ceará, Brazil
5Reference Center at Children’s Cancer Diagnosis and Adolescents Dr. Murilo Martins, Albert Sabin Hospital, Ceará, Brazil.
*Corresponding author
E-mail: acomoreira@unifor.br
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