U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

National Guideline Alliance (UK). Pancreatic cancer in adults: diagnosis and management. London: National Institute for Health and Care Excellence (NICE); 2018 Feb. (NICE Guideline, No. 85.)

Cover of Pancreatic cancer in adults: diagnosis and management

Pancreatic cancer in adults: diagnosis and management.

Show details

5Diagnosis

5.1. People with jaundice

Review question: What is the most effective diagnostic pathway (imaging +/-CA 19–9, biopsy (cytology or histology)) for adults with suspected pancreatic cancer in secondary care who have jaundice?

5.1.1. Introduction

Obstructive jaundice is the most common presenting symptom in people with pancreatic cancer, although it is to be noted that most people presenting with jaundice do not actually have pancreatic cancer.

There is currently uncertainty about the most accurate technique for diagnosing the disease in people with obstructive jaundice. CT scans are commonly used to diagnose pancreatic cancer in this group of people, however it is not always possible for the CT scan to visualise the cancer that is causing the obstruction. Ultrasound is another technique which can identify pancreatic cancer. MRI and fluorodeoxyglucose-positron emission tomography/CT (FDG-PET/CT) are both increasingly being used but their diagnostic accuracy in this group of people is not clearly understood. Whether histology and cytology are needed to make the diagnosis of pancreatic cancer in someone with obstructive jaundice is uncertain, with some centres operating on imaging alone. There is also variation in practice as to how the histology and cytology are obtained. The role of cancer antigen 10-9 (CA 19-9) in combination with imaging is not defined.

In the group of people thought not suitable for resection based on imaging, brushing the duct (for cytology) at the time of ERCP and stenting is common. Where this does not confirm a diagnosis, endoscopic ultrasound (EUS) and fine needle aspiration (FNA) is usually done. However there are still a small group of people in whom the imaging is highly suggestive of malignancy but the cytology/histology does not confirm, leaving the question of what to do next.

Guidance is needed on the most effective diagnostic pathway to identify pancreatic cancer in people who have jaundice.

5.1.1.1. Review protocol summary

The review protocol summary used for this question can be found in Table 17. Full details of the review protocol can be found in Appendix C.

Table 17. Clinical review protocol summary for the review of most effective diagnostic pathway for people with suspected pancreatic cancer who have jaundice.

Table 17

Clinical review protocol summary for the review of most effective diagnostic pathway for people with suspected pancreatic cancer who have jaundice.

5.1.2. Description of Clinical Evidence

Six observational studies (n=806) - 1 multicentre prospective cohort study (n=159) and five single-centre retrospective cohort studies (n=647) - were included in the review. A summary of the included studies is presented in Table 18.

One study (n=47) reported on the diagnostic accuracy of spiral CT. This study was carried out in the USA and included patients with obstructive jaundice with a suspicion of pancreatic cancer (Agarwal et al. 2004).

One study (n=47) reported on the diagnostic accuracy of EUS. This study was carried out in the USA and included patients with obstructive jaundice with a suspicion of pancreatic cancer (Agarwal et al. 2004).

Five studies (n=691) reported on the diagnostic accuracy of EUS-FNA based cytology (Agarwal et al. 2004; Kim et al. 2015; Oppong et al. 2010; Ross et al. 2008; Tummala et al. 2013). All studies included patients with obstructive jaundice with a suspicion of pancreatic cancer. One study was conducted in the UK (Oppong et al. 2010), whilst the remaining 4 studies were conducted in the USA.

One prospective multicentre UK study (n=393) – known as PET-PANC - reported on the diagnostic accuracy of MDCT and FDG-PET/CT (Ghaneh et al. 2018) in patients with obstructive jaundice and a suspicion of pancreatic cancer. The main aim of the latter study was to assess - in a multicentre setting and using a standardised protocol - whether the addition of FDG-PET/CT to MDCT, which is standard practice in the UK, provides tangible diagnostic and staging benefits. Two studies (n=89) reported on the diagnostic accuracy of ERCP + brushings of biliary strictures (Oppong et al. 2010; Ross et al. 2008). Both studies included patients with obstructive jaundice with a suspicion of pancreatic. One study was conducted in the UK (Oppong et al. 2010), with the other study conducted in the USA (Ross et al. 2008).

All included studies reported on diagnostic accuracy outcome measures, whilst only 2 studies reported adverse effects or complications. Positive and likelihood ratios were calculated, where appropriate, from the raw diagnostic test accuracy data or the estimated sensitivity and specificity of the studies to enable evaluation of the relevant tests. The QUADAS-2 checklist was used to evaluate the risk of bias and indirectness (applicability) of the studies.

Further information about the search strategy can be found in Appendix D. See study selection flow chart in Appendix E, single and multiple test ROC curves and forest plots in Appendix H, summary of Risk of Bias in Appendix J, study evidence tables in Appendix F and list of excluded studies in Appendix G.

5.1.3. Summary of included studies

A summary of the studies that were included in this review is presented in Table 18.

Table 18. Summary of included studies.

Table 18

Summary of included studies.

5.1.4. Clinical evidence profile

The clinical evidence profiles for this review question are presented in Table 19 to Table 22.

Table 19. Summary of clinical evidence for CT to detect malignancy in people with jaundice.

Table 19

Summary of clinical evidence for CT to detect malignancy in people with jaundice.

Table 20. Summary of clinical evidence for EUS to detect malignancy in people with jaundice.

Table 20

Summary of clinical evidence for EUS to detect malignancy in people with jaundice.

Table 21. Summary of clinical evidence for EUS-FNA cytology to detect malignancy in people with jaundice.

Table 21

Summary of clinical evidence for EUS-FNA cytology to detect malignancy in people with jaundice.

Table 22. Summary of clinical evidence for ERCP + brushings of biliary strictures to detect malignancy in people with jaundice.

Table 22

Summary of clinical evidence for ERCP + brushings of biliary strictures to detect malignancy in people with jaundice.

Table 23. Summary of clinical evidence for FDG-PET/CT to detect malignancy in people with jaundice.

Table 23

Summary of clinical evidence for FDG-PET/CT to detect malignancy in people with jaundice.

5.1.5. Economic evidence

A literature review of published cost effectiveness analyses did not identify any relevant studies for this topic. Although there were potential implications for resource use associated with making recommendations in this area, other topics in the guideline were agreed as a higher economic priority. Consequently, bespoke economic modelling was not done for this topic.

5.1.6. Evidence Statements

5.1.6.1. Computed tomography (CT)

Diagnostic accuracy

Low quality evidence from 1 retrospective cohort study (n=47) found that spiral CT (n=47) had a low sensitivity of 0.67 (95% CI, 0.51-0.8) and a high specificity of 1.0 (95% CI, 0.16-1.0) in detecting malignancy in pancreatic cancer patients with obstructive jaundice. The positive likelihood ratio of 3.98 (95% CI, 0.31-50.34) suggests that a positive result for malignancy is not particularly useful for ruling it in, though there is uncertainty in the estimate. The negative likelihood ratio of 0.33 (95% CI, 0.22-0.50) suggests that a negative result for malignancy is not particularly useful for ruling it out.

Moderate quality evidence from 1 prospective cohort study (n=148) found that multidetector computed tomography had a high sensitivity of 0.9 (95% CI, 0.82-0.95) and a low specificity of 0.58 (95% CI, 0.44-0.71) in detecting malignancy in pancreatic cancer patients with obstructive jaundice. The positive likelihood ratio of 2.14 (95% CI, 1.57-2.92) suggests that a positive result for malignancy is not particularly useful for ruling it in. The negative likelihood ratio of 0.17 (95% CI, 0.09-0.33) suggests that a negative result for malignancy is moderately useful for ruling it out, though there is substantial uncertainty in the estimate.

Adverse events

In 1 multicentre prospective cohort study (n=583) that examined the diagnostic test accuracy of CT, no adverse events related to the tests were reported.

5.1.6.2. Endoscopic ultrasonography (EUS)

5.1.6.2.1. EUS
Diagnostic accuracy

Moderate quality evidence from 1 retrospective observational study (n=47) people found that EUS had high sensitivity of 1.0 (95% CI, 0.92-1.0) and low specificity of 0.5 (95%CI, 0.01-0.99) in detecting malignancy in pancreatic cancer patients with obstructive jaundice. The positive likelihood ratio of 2.0 (95% CI, 0.5-8.0) suggests that a positive result for malignancy is not particularly useful for ruling it in, though there is uncertainty in the estimate. The negative likelihood ratio of 0 suggests that a negative result for malignancy is very useful for ruling it out.

Adverse events

No evidence was identified to inform this outcome.

5.1.6.2.2. EUS-FNA cytology
Diagnostic accuracy

Low quality evidence from a meta-analysis of 5 retrospective observational studies (n=691) found that EUS-FNA-based cytology had a moderate sensitivity of 0.85 (95% CI, 0.79-0.9) and a high specificity of 0.96 (95% CI, 0.86-0.99) in detecting malignancy in pancreatic cancer patients with obstructive jaundice. The positive likelihood ratio of 22.2 (95% CI, 5.81-84.75) suggests that a positive result for malignancy is very useful for ruling it in, though there is uncertainty in the estimate. The negative likelihood ratio of 0.15 (95% CI, 0.11-0.22) suggests that a negative result for malignancy is moderately useful for ruling it out, though there is uncertainty in the estimate.

Adverse events

Low quality evidence from 1 retrospective observational study (n=342 with resectable pancreatic cancer) found that there were 2 overall complications related to the EUS-FNA procedure: 1 patient had acute pancreatitis requiring hospitalization for 3 days and another patient had aspiration pneumonia requiring oral antibiotics.

5.1.6.3. Endoscopic retrograde cholangiopancreatography (ERCP)

5.1.6.3.1. ERCP + Brushings of biliary strictures
Diagnostic accuracy

Very low quality evidence from 2 retrospective observational studies with (n=39; n=50) found that ERCP plus brushings of biliary strictures had a low sensitivity, ranging from 0.13 to 0.65 and a high specificity of 1.0 (in both studies) in detecting malignancy in pancreatic cancer patients with obstructive jaundice. The positive likelihood ratios ranged from 7.71 (95% CI, 0.54-110.87) to 6.1 (95% CI, 0.35-107.4) suggesting that a positive result for malignancy is moderately useful for ruling it in, though there is uncertainty in the estimates. The negative likelihood ratios ranged from 0.35 (95% CI, 0.22-0.56) to 0.87 (95% CI, 0.75-1.0) suggesting that a negative result for malignancy is not particularly useful for ruling it out.

Adverse events

No evidence was identified to inform this outcome.

5.1.6.4. Positron emission tomography/-CT (PET/-CT)

Diagnostic accuracy

Moderate quality evidence from 1 prospective cohort study (n=148) found that FDG-PET/CT had a high sensitivity of 0.96 (95% CI, 0.89-0.99) and a low specificity of 0.53 (95% CI, 0.39-0.66) in detecting malignancy in pancreatic cancer patients with obstructive jaundice. The positive likelihood ratio of 2.02 (95% CI, 1.53-2.66) suggests that a positive result for malignancy is not particularly useful for ruling it in. The negative likelihood ratio of 0.08 (95% CI, 0.03-0.22) suggests that a negative result for malignancy is very useful for ruling it out, though there is substantial uncertainty in the estimate.

Adverse events

In 1 multicentre prospective cohort study (n=583) that examined the diagnostic test accuracy of CT, no adverse events related to the tests were reported.

5.1.7. Recommendations

1.

For people with obstructive jaundice and suspected pancreatic cancer, offer a pancreatic protocol CT scan before draining the bile duct.

2.

If the diagnosis is still unclear, offer fluorodeoxyglucose-positron emission tomography/CT (FDG-PET/CT) and/or endoscopic ultrasound (EUS) with EUS-guided tissue sampling.

3.

Take a biliary brushing for cytology if:

  • endoscopic retrograde cholangiopancreatography (ERCP) is being used to relieve the biliary obstruction and
  • there is no tissue diagnosis.

5.1.8. Evidence to recommendations

5.1.8.1. Relative value placed on the outcomes considered

Diagnostic accuracy (sensitivity, specificity, positive predictive value and negative predictive value) and adverse events were considered the critical outcomes for this question. Diagnostic accuracy was reported for all comparisons of interest. Adverse events were only reported for EUS-FNA, MDCT and FDG-PET/CT.

5.1.8.2. Quality of evidence

Evidence was identified on the diagnostic accuracy of spiral and MDCT, EUS, EUS-FNA cytology, FDG-PET/CT and ERCP plus brushings of biliary strictures. The quality of the evidence for FDG-PET/CT was moderate, for ERCP plus brushings of biliary strictures it ranged from very low to low, for CT was low (for spiral CT) to moderate (for MDCT), EUS-FNA cytology was low and for EUS was moderate.

The committee noted that all studies, except for FDG-PET/CT, had either a serious or a very serious risk of bias due to different reference standards being used across the study sample; a lack of blinding; the test being evaluated being included in the reference standard (potentially leading to an overestimation of test accuracy); people inappropriately excluded from the analysis. The committee had more confidence in the quality of evidence from the report related to FDG-PET/CT by Ghaneh et al. (2018) because it was the largest multicentre study, it was conducted in a UK NHS setting (and therefore directly applicable) and the study design was judged by the committee to be more robust than that of the other included studies. Therefore in their discussion the committee placed relatively more weight on the evidence from this study than in the rest of the evidence base.

The committee also noted that all patients had either imaging or ERCP in order to get into these studies –the quality of this imaging could have had an effect on the accuracy results. In addition the data for spiral CT were very old as the paper was from 2004. The committee considered that the accuracy of CT was likely to be better than reported by these data as the technology has advanced significantly since that time, as suggested by the data for MDCT. They also agreed that CT was able to image the entire body which would be beneficial in these patients and this contributed to the committee’s decision to make a strong recommendation..

The committee noted that adverse event data were only found for EUS-FNA, CT and FDG-PET/CT. Based on their clinical knowledge and experience, that there is a relatively low occurrence of adverse events with these procedures, the committee did not apply much weight to this data when making recommendations.

No evidence was found on the diagnostic accuracy of CA19-9 or CT-guided biopsy in diagnosing pancreatic cancer in people with jaundice. Therefore no recommendations were made about these investigations. No further research was recommended since these were not considered high priorities for research funding.

5.1.8.3. Consideration of clinical benefits and harms

The evidence showed that there was heterogeneity in results for CT with one study reporting high specificity for detecting pancreatic cancer but low sensitivity whereas the other study reported the opposite findings (high sensitivity and lower specificity). The study with higher sensitivity and lower specificity provided higher quality evidence and the committee gave more weight to this in their discussion. EUS had low specificity but high sensitivity. Based on their clinical experience and knowledge the committee noted that a CT scan was a less invasive technique and was able to identify metastases, which EUS could not do. Given that CT is less invasive and would capture most positive cases (according to the higher quality evidence) the committee therefore recommended CT as the first investigation to diagnose pancreatic cancer as a rule-out test in someone with obstructive jaundice. Based on their clinical knowledge and experience, the committee noted that if a CT scan is used, a pancreatic protocol CT scan should be used to ensure good visualisation of any pathology in the pancreas. The committee noted that this is current practice and that their recommendation reinforces this message. They also agreed, based on their knowledge and experience, that if biliary drainage was performed to relieve the jaundice before the CT scan was conducted, this would detrimentally affect the interpretation of the CT scan. They therefore agreed that the CT scan should be conducted before biliary drainage.

For people with uncertain findings after CT scanning had been conducted, the committee believed that FDG-PET/CT added significant additional information. Based on the evidence and their knowledge committee members noted that this was particularly the case in in the detection of metastatic disease. In addition, due to its non-invasive nature and the low false negative rates FDG-PET/CT was considered to be an appropriate additional diagnostic test to rule out malignancy in people with suspected pancreatic cancer. The committee recommended FDG-PET/CT and / or EUS with tissue sampling. If EUS is used in combination with FDG-PET/CT or on its own, taking a tissue sample at the same time as EUS is recommended because it would be needed to confirm the diagnosis and taking it at the same time as EUS would reduce the need for repeated tests which would be more acceptable to patients. The committee noted that EUS with tissue sampling had both high sensitivity and specificity whereas FDG-PET/CT had high sensitivity but lower specificity. The committee decided that the non-invasive nature of FDG-PET/CT, the low false negative rate and the additional information related to metastic disease that it can provide, would put FDG-PET/CT alongside EUS with tissue sampling as the next step if further diagnostic information is required after the CT scan. The committee therefore decided that a FDG-PET/CT scan should be conducted and / or EUS (with tissue sampling) if the diagnosis is still unclear after CT.

The committee noted that the evidence for ERCP plus brushings of biliary strictures showed high specificity but relatively low sensitivity and was of very low or low quality. They therefore agreed not to make any recommendation about whether ERCP should be performed or not. However, the committee noted, based on their knowledge and experience, that some people who are deeply jaundiced or who are unfit for surgery will have an ERCP to relieve the obstruction that is causing the jaundice before they have a tissue diagnosis. Brushings of biliary strictures taken during the ERCP will give further diagnostic information which will inform treatment. They therefore agreed to recommend biliary brushing to obtain cytology if an ERCP is being performed and there is no tissue diagnosis. The committee agreed that despite the low quality of the evidence, this should be a strong recommendation because having the diagnostic information provided by the brushings was essential, and in this group it could only be obtained by biliary brushings.

The potential benefits of the recommendations made were considered to be a more efficient pathway to diagnosis for people with obstructive jaundice which optimises non-invasive investigations and a reduction in the need for multiple diagnostic investigations. The potential harms were complications associated with the use of EUS and ERCP. However, as these complication rates are low the potential benefits were considered to outweigh the potential harms.

5.1.8.4. Consideration of economic benefits and harms

The committee noted that whilst no relevant published economic evaluations had been indentified, diagnosis (including patients with jaundice) formed part of the diagnosis and staging pathway for the cost utility analysis in a health technology assessment (HTA) by Ghaneh et al. (2018) discussed in detail in section 7.5.1.

The HTA highlighted that including FDG-PET/CT as part of the diagnostic and staging work up of patients with suspected pancreatic cancer was very likely cost saving and health improving. It was acknowledged that the HTA did not look at the cost effectiveness of the addition of FDG-PET/CT in a sub-group of patients with obstructive jaundice although this group was part of the larger study cohort considered. However, the committee could not see any clinical reason why the conclusions would not remain the same if such a sub group was considered.

The committee also acknowledged that the majority of the cost savings and health improvements identified in the HTA would be as a result of better staging and a reduction in unnecessary resections (which is why the study was discussed in detail for the staging topic). The recommendations for that topic, for diagnosis of people with obstructive jaundice and suspected pancreatic cancer, and staging of those with confirmed pancreatic cancer almost identically match the diagnosis and staging pathway used as the intervention in the HTA’s cost utility study. The committee therefore considered the reasons discussed in section 7.8.4. applied to the diagnostic recommendation for FDG-PET/CT as well (see recommendation 2). The committee were therefore confident this recommendation was cost effective and very likely cost saving and health improving.

The recommendation would lead to an initial increase in patients with obstructive jaundice receiving FDG-PET/CT as only a minority of this patient group currently receive these. Given the relatively large patient group this could be significant. The HTA strongly suggests that this initial increase in resource use would be recoupedwithin a year.

5.1.9. References

  • Agarwal B, Abu-Hamda E, Molke KL et al. (2004) Endoscopic ultrasound-guided fine needle aspiration and multidetector spiral CT in the diagnosis of pancreatic cancer. American Journal of Gastroenterology 99(5): 844–50 [PubMed: 15128348]
  • Ghaneh P, Hanson R, Titman A et al. (2018) PET-PANC: multicentre prospective diagnostic accuracy and health economic analysis study of the impact of combined modality 18fluorine-2-fluoro-2-deoxy-d-glucose positron emission tomography with computed tomography scanning in the diagnosis and management of pancreatic cancer. Health Technology Assessment 22(7) [PMC free article: PMC5817411] [PubMed: 29402376]
  • Kim JJ, Walia S, Lee SH et al. (2015) Lower yield of endoscopic ultrasound-guided fine-needle aspiration in patients with pancreatic head mass with a biliary stent. Digestive diseases and sciences 60(2): 543–549 [PubMed: 25245115]
  • Oppong K, Raine D, Nayar M et al. (2010) EUS-FNA versus biliary brushings and assessment of simultaneous performance in jaundiced patients with suspected malignant obstruction. Journal of the Pancreas 11(6): 560–567 [PubMed: 21068487]
  • Ross WA, Wasan SM, Evans DB et al. (2008) Combined EUS with FNA and ERCP for the evaluation of patients with obstructive jaundice from presumed pancreatic malignancy. Gastrointestinal endoscopy 68(3): 461–466 [PubMed: 18384788]
  • Tummala P, Munigala S, Eloubeidi MA et al. (2013) Patients with obstructive jaundice and biliary stricture±mass lesion on imaging: prevalence of malignancy and potential role of EUS-FNA. Journal of clinical gastroenterology 47(6): 532–537 [PubMed: 23340062]

5.2. People without jaundice but with a pancreatic abnormality

Review question: What is the most effective diagnostic pathway (imaging +/-CA 19–9, biopsy (cytology or histology)) for adults with suspected pancreatic cancer in secondary care who do not have jaundice but have a pancreatic abnormality on imaging?

5.2.1. Introduction

The availability and use of imaging, both ultrasound and CT, continues to increase in clinical practice and, as a consequence, incidental lesions are detected with increasing frequency. Incidental lesions in the pancreas, both solid and cystic, in asymptomatic people are a common finding. There is no consensus as to the most appropriate pathway to establish an accurate diagnosis in this patient group.

Pancreatic CT scanning is regarded as the mainstay of the imaging pathway, but the role of pancreatic MRI and FDG-PET/CT, although not well defined, is increasing.

In addition, the role of both cytology and histology and the best method of obtaining tissue to confirm the diagnosis has not been established. Imaging may also reveal metastatic disease, which could be sampled to help establish the diagnosis.

Guidance is needed on the most effective diagnostic pathway to identify pancreatic cancer in people who have a pancreatic abnormality on imaging.

5.2.1.1. Review protocol summary

The review protocol summary used for this question can be found in Table 24. Full details of the review protocol can be found in Appendix C.

Table 24. Clinical review protocol summary for the review of the most effective diagnostic pathway for people with suspected pancreatic cancer who do not have jaundice but have a pancreatic abnormality on imaging.

Table 24

Clinical review protocol summary for the review of the most effective diagnostic pathway for people with suspected pancreatic cancer who do not have jaundice but have a pancreatic abnormality on imaging.

5.2.2. Description of clinical evidence

Twenty-one articles reporting a total of 32 datasets were identified: 3 of these were RCTs (Bang et al. 2012; Lee et al. 2014; Ramesh et al. 2015), 13 were prospective cohort studies (Bournet et al. 2015; Bournet et al. 2009; Fabbri et al. 2011; Harewood & Wiersema 2002; Iglesias-Garcia et al. 2007; Kliment et al. 2010; Krishna et al. 2009; Mishra et al. 2006; Seicean et al. 2016; Strand et al. 2014; Touchefeu et al. 2009; Wakatsuki et al. 2005; Wittman et al. 2006) and 5 were retrospective cohort studies (Fritscher-Ravens et al. 2002; Hikichi et al. 2009; Tamm et al. 2007; Yang et al. 2015; Yusuf et al. 2009). A summary of the included studies is presented in Table 25.

The majority of the studies examined the diagnostic test accuracy of EUS-FNA for detecting malignancy in patients with suspected pancreatic cancer due to a solid lesion identified through previous imaging (e.g. EUS, CT, MRI, ERCP). The majority of the studies reported sensitivity and specificity, as well as positive/negative predictive value. Three articles (Hikichi et al. 2009; Ramesh et al. 2015; Yusuf et al. 2009) contributed two sets of data to the review on EUS-FNA. The majority of the studies also used a composite ‘gold standard’ reference test generally comprised of histo-/cyto-pathology from surgery, and subsequent clinical and imaging follow-up results. The majority of the studies also reported that there were no procedure-related adverse events, serious or otherwise. No studies were found that examined percutaneous liver biopsy, laparoscopy + biopsy.

One single centre retrospective cohort study (n=117) examined the diagnostic accuracy of multidetector CT (Tamm et al. 2007) in detecting malignancy in solid lesions initially identified through imaging.

Two single centre cohort studies (n=330) – 1 prospective (n=213; Krishna et al. 2009) and 1 retrospective (n=117; Tamm et al. 2007) - examined the diagnostic accuracy of EUS in detecting malignancy in solid lesions initially identified through imaging. The sample in Krishna et al. (2009) had a low prevalence of malignant lesions (0.52) and included 15% patients whose lesions were revealed to be cystic by EUS-FNA.

Twenty-two datasets (n=2869) from 19 studies - 3 RCTs (Bang et al. 2012; Lee et al. 2014; Ramesh et al. 2015) and 16 (11 prospective and 5 retrospective) cohort studies - examined the diagnostic accuracy of EUS-FNA in detecting malignancy in solid lesions initially identified through imaging (Bournet et al. 2009, 2015; Fabbri et al. 2011; Fritscher-Ravens et al. 2002; Harewood & Wiersema 2002; Hikichi et al. 2009; Iglesias-Garcia et al. 2007; Kliment et al. 2010; Krishna et al. 2009; Mishra et al. 2006; Seicean et al. 2016; Tamm et al. 2007; Touchefeu et al. 2009; Wakatsuki et al. 2005; Wittman et al. 2006; Yusuf et al. 2009). The majority of these studies used a 22-gauge needle to extract a cytological specimen. The number of included studies (≥4) allowed a meta-analysis of the diagnostic test accuracy data to be performed, which produces a summary point estimate of the sensitivity and specificity of EUS-FNA. Although there was not sufficient data to examine heterogeneity for covariates such as needle type and type of reference test, a subgroup analysis by type of study (RCT/prospective cohort vs retrospective cohort) was conducted.

Four studies (n=158) - 2 RCTs (Bang et al. 2012; Lee et al. 2014) and 2 prospective cohort studies (Strand et al. 2014; Wittman et al. 2006) - examined the diagnostic accuracy of EUS-core biopsy in detecting malignancy in solid lesions initially identified through imaging. The number of included studies (≥4) allowed a meta-analysis of the diagnostic test accuracy data to be performed, which produces a summary point estimate of the sensitivity and specificity of EUS-core biopsy. The two RCTs, which randomised participants to receive either EUS-FNA or EUS-core, both used fine biopsy (ProCore) needles (EUS-FNB), whilst the cohort studies used either FNB (Strand et al. 2014) or trucut (Wittman et al. 2006) biopsy needles (EUS-TNB).

One prospective cohort study (n=36) examined the diagnostic accuracy of combining EUS-FNA with EUS-Core (Wittman et al. 2006).

One multicentre retrospective cohort study (n=60) examined the diagnostic accuracy of percutaneous US-guided core in detecting malignancy in solid lesions initially identified through imaging (Yang et al. 2015).

One multicentre retrospective cohort study (n=15) examined the diagnostic accuracy of percutaneous US-guided FNA + core in detecting malignancy in solid lesions initially identified through imaging (Yang et al. 2015).

Positive and likelihood ratios were calculated, where appropriate, from the raw diagnostic test accuracy data or the estimated sensitivity and specificity of the studies to enable evaluation of the relevant tests. The QUADAS-2 checklist was used to evaluate the risk of bias and indirectness (applicability) of the studies.

5.2.3. Summary of included studies

A summary of the studies that were included in this review is presented in Table 25.

Table 25. Summary of included studies.

Table 25

Summary of included studies.

5.2.4. Clinical evidence profile

The clinical evidence profiles for this review question are presented in Table 26 to Table 33.

5.2.4.1. Computed tomography

Table 26. Summary of clinical evidence for computed tomography to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Table 26

Summary of clinical evidence for computed tomography to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

5.2.4.2. Endoscopic ultrasonography (EUS)

5.2.4.2.1. EUS
Table 27. Summary of clinical evidence for EUS to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Table 27

Summary of clinical evidence for EUS to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

5.2.4.2.2. EUS-FNA
Table 28. Summary of clinical evidence for EUS-FNA to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Table 28

Summary of clinical evidence for EUS-FNA to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Table 29. Pooled sensitivity and specificity of EUS-FNA by type of study.

Table 29

Pooled sensitivity and specificity of EUS-FNA by type of study.

5.2.4.2.3. EUS-Core (FNB or TNB)
Table 30. Summary of clinical evidence for EUS-guided core biopsy (FNB or trucut) to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Table 30

Summary of clinical evidence for EUS-guided core biopsy (FNB or trucut) to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

5.2.4.2.4. EUS-FNA + Core
Table 31. Summary of clinical evidence for EUS-FNA + Core to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Table 31

Summary of clinical evidence for EUS-FNA + Core to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Percutaneous ultrasonography
5.2.4.2.5. Percutaneous US-guided Core
Table 32. Summary of clinical evidence for percutaneous US-guided core to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Table 32

Summary of clinical evidence for percutaneous US-guided core to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

5.2.4.2.6. Percutaneous US-guided FNA + Core
Table 33. Summary of clinical evidence for percutaneous US-guided FNA + core to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

Table 33

Summary of clinical evidence for percutaneous US-guided FNA + core to detect malignancy in people without jaundice but who have a pancreatic abnormality on imaging.

5.2.5. Economic evidence

A literature review of published cost effectiveness analyses did not identify any relevant studies for this topic. Although there were potential implications for resource use associated with making recommendations in this area, other topics in the guideline were agreed as a higher economic priority. Consequently, bespoke economic modelling was not done for this topic.

5.2.6. Evidence statements

5.2.6.1. Computed tomography (CT)

Diagnostic accuracy

Moderate quality evidence from 1 single centre retrospective cohort study (n=117) found that multidetector CT had a high sensitivity of 0.97 (95% CI, 0.91-0.99) and a low specificity of 0.72 (95% CI, 0.46-0.89) in detecting malignant incidental solid pancreatic lesions in adults with suspected pancreatic cancer. The positive likelihood ratio of 3.49 (1.66-7.36) suggests that a positive result for malignancy is not particularly useful for ruling it in, though there is uncertainty in the estimate. The negative likelihood ratio of 0.04 (95% CI, 0.01-0.13) suggests that a negative result for malignancy is very useful for ruling it out, though there is uncertainty in the estimate.

Adverse events

No evidence was identified to inform this outcome.

5.2.6.2. Endoscopic ultrasonography (EUS)

5.2.6.2.1. EUS
Diagnostic accuracy

Moderate quality evidence from 2 single centre cohort studies - 1 prospective (n=213) and 1 retrospective (n=117) - found that EUS had a high sensitivity ranging from 0.99 to 1.0 and low specificity ranging from 0.5 to 0.66 in detecting malignant incidental solid pancreatic lesions in adults with suspected pancreatic cancer. The positive likelihood ratios were 1.98 (95% CI, 1.25-3.14) and 2.94 (95% CI, 2.25-3.85) suggesting that a positive result for malignancy is not useful for ruling it in. The negative likelihood ratios were 0 and 0.02 (95% CI, 0-0.15) suggesting that a negative result for malignancy is very useful for ruling it out, though there is uncertainty in the latter estimate.

Adverse events

No evidence was identified to inform this outcome.

5.2.6.2.2. EUS-FNA
Diagnostic accuracy

Moderate quality evidence from a meta-analysis of 22 studies (n=2869) found that endoscopic ultrasound fine needle aspiration had a moderate pooled sensitivity of 0.89 (95% CI, 0.85-0.92) and a high pooled specificity of 0.99 (95% CI, 0.96-1.0) in detecting malignant incidental solid pancreatic lesions in adults with suspected pancreatic cancer. The positive likelihood ratio of 121.03 (95%, 20.64-709.55) suggests that a positive result for malignancy is very useful for ruling it in. The negative likelihood ratio of 0.11 (0.08-0.15) suggests that a negative result for malignancy is moderately useful for ruling it out, though there is uncertainty in the estimate.

A subgroup analysis by study type (RCTs and prospective cohort studies vs retrospective cohort studies) showed that there was no significant difference between the two groups in the estimated pooled sensitivity (0.89 [95% CI, 0.84-0.93] vs 0.88 [95% CI, 0.84-0.91], respectively) and pooled specificity (0.99 [95% CI, 0.91-1.0] vs 0.99 [95% CI, 0.97-1.0], respectively), although there was more uncertainty in the pooled estimates from the RCT/prospective cohort study group. The similar positive likelihood ratios of 92.82 (95% CI, 9.29-927.71) and 109.95 (95% CI, 25.14-480.83) in the two subgroups support the conclusion above that a positive result for malignancy is very useful for ruling it in. Similarly, the negative likelihood ratios for the subgroups of 0.11 (95% CI, 0.07-0.17) and 0.12 (95% CI, 0.09-0.16) also support the conclusion above that a negative result for malignancy is moderately useful for ruling it out, though there is uncertainty in the estimates.

Adverse events

Fourteen studies (N=2123) reported data on adverse events with complication rates ranging from 0% to 4%. Nine studies reported that there were no adverse events, whilst the most common adverse event reported in the remaining 8 studies was mild pancreatitis (13 reported cases). Other reported adverse events included post-procedural pain (2 cases), bleeding and fever (1 case each).

5.2.6.2.3. EUS-Core (FNB or trucut)
Diagnostic accuracy

Very low quality evidence from a meta-analysis of 4 studies (n=154) found that endoscopic ultrasound core biopsy had a low pooled sensitivity of 0.7 (95% CI, 0.3-0.93) and a high pooled specificity of 0.99 (95% CI, 0.03-1.0) in detecting malignant incidental solid pancreatic lesions in adults with suspected pancreatic cancer. The positive likelihood ratio of 176.61 (95% CI, 0.02-1867693) suggests that a positive result for malignancy is very useful for ruling it in, though there is substantial uncertainty in the estimate. The negative likelihood ratio of 0.3 (95% CI, 0.09-1.02) suggests that a negative result for malignancy is not particularly useful for ruling, though there is substantial uncertainty in the estimate.

Adverse events

The studies reported no serious procedure-related adverse events. The complication rate ranged from 0% to 5.2%. One study reported a case of mild acute pancreatitis that required hospitalisation for 2 days, and 1 study reported 2 cases of gastric haematoma and 1 case of mild bleeding.

5.2.6.2.4. EUS-FNA + Core
Diagnostic accuracy

Low quality evidence from 1 single-centre prospective cohort study (N=36) found that combining EUS-FNA with EUS-Core biopsy had a moderate sensitivity of 0.76 (95% CI, 0.55-0.91) and a high specificity of 1.0 (95% CI, 0.72-1.0) in detecting malignant incidental solid pancreatic lesions in adults with suspected pancreatic cancer. The positive likelihood ratio of 18 (95% CI, 1.18-273.95) suggests that a positive result for malignancy is very useful for ruling it in, though there is substantial uncertainty in the estimate. The negative likelihood ratio of 0.24 (95% CI, 0.12-0.48) suggests that a negative result for malignancy is not particularly useful for ruling it out, though there is uncertainty in the estimate.

Adverse events

The study did not report any serious adverse events. There was a 3% complication rate with 1 case of moderate self-limiting abdominal pain (not requiring analgesia) after biopsy of a pancreatic tail lesion.

5.2.6.3. Percutaneous ultrasonography (percutaneous US)

5.2.6.3.1. Percutaneous US-guided Core
Diagnostic accuracy

Low quality evidence from 1 multicentre retrospective cohort study (n=60) found that percutaneous US-guided core biopsy had a high sensitivity of 0.93 (95% CI, 0.82-0.98) and a high specificity of 1.0 (95% CI, 0.54-1.0) in detecting malignant incidental solid lesions in adults with suspected pancreatic cancer. The positive likelihood ratio of 12.85 (95% CI, 0.89-186.03) suggests that a positive result for malignancy is very useful for ruling it in, though there is substantial uncertainty in the estimates. The negative likelihood ratio of 0.07 (95% CI, 0.03-0.19) suggests that a negative result for malignancy is very useful for ruling it out, though there is uncertainty in the estimates.

Adverse events

The study did not report any serious adverse events. There was a 3% complication rate with 1 case of haematoma and 1 case of pain, both reported immediately after the biopsy was taken.

5.2.6.3.2. Percutaneous US-guided FNA + Core
Diagnostic accuracy

Low quality evidence from 1 multicentre retrospective cohort study (n=15) found that percutaneous US-guided core biopsy combined with PUS-FNA had high sensitivity of 0.92 (95% CI, 0.64-1.0) and a high specificity of 1.0 (95% CI, 0.16-1.0) in detecting malignant incidental solid lesions in adults with suspected pancreatic cancer. The positive likelihood ratio of 5.36 (95% CI, 0.42-67.71) suggests that a positive result for malignancy is moderately useful for ruling it in, though there is substantial uncertainty in the estimates. The negative likelihood ratio of 0.08 (95% CI, 0.01-0.51) suggests that a negative result for malignancy is very useful for ruling it out, though there is substantial uncertainty in the estimates.

Adverse events

The study did not report any serious adverse events. There was a complication rate of 7% with 1 case of pain reported immediately after the biopsy was taken.

5.2.7. Recommendations

4.

Offer a pancreatic protocol CT scan to people with pancreatic abnormalities but no jaundice.

5.

If the diagnosis is still unclear, offer fluorodeoxyglucose-positron emission tomography/CT (FDG-PET/CT) and/or EUS with EUS-guided tissue sampling.

6.

If cytological or histological samples are needed, offer EUS with EUS-guided tissue sampling.

5.2.8. Evidence to recommendations

5.2.8.1. Relative value placed on the outcomes considered

Diagnostic accuracy (sensitivity, specificity, positive predictive value and negative predictive value) and adverse events were considered the critical outcomes for this question. Diagnostic accuracy was reported for all interventions of interest. Adverse events were reported for all interventions except CT and EUS.

5.2.8.2. Quality of evidence

Evidence was identified on the diagnostic accuracy of CT, EUS, EUS-FNA, EUS-core, EUS-FNA + core, percutaneous US-guided core and percutaneous US-guided FNA + core. The quality of the evidence for CT and EUS-FNA was moderate, for EUS was high, for all other investigations was either very low or low.

Given the low quality of the data for EUS-core, EUS-FNA + core, percutaneous US-guided core and percutaneous US-guided FNA + core, the committee were less certain of the balance between diagnostic accuracy and potential adverse events for these investigations. They, therefore, agreed to apply more weight to the investigations with moderate and high quality data. They did not make any recommendations about core biopsy by percutaneous routes.

No evidence was identified on percutaneous liver or pancreatic biopsy or laparoscopy + biopsy. Therefore, no recommendations were made about these investigations. No further research was recommended since these were not considered high priorities for research funding.

5.2.8.3. Consideration of clinical benefits and harms

The committee noted that of the investigations with moderate or high quality evidence, EUS had shown the highest sensitivity but the lowest specificity for diagnosing malignancy in a solid lesion suspected to be pancreatic cancer. Given that other investigations had similar sensitivities but better specificities, they agreed not to make a recommendation about EUS alone.

The committee noted, based on the evidence, that whilst the positive likelihood ratio for CT was not as good as that for EUS-FNA/FNB, CT had a better negative likelihood ratio. They also agreed, based on their knowledge and experience, that CT was more widely available than EUS-FNA and was non-invasive so the risk of adverse events was lower. Therefore, they agreed to recommend a CT scan as the first option in people with a solid lesion suspected to be pancreatic cancer as a ruling out test. Based on their clinical knowledge and experience, the committee noted that if a CT scan is used a pancreatic protocol CT scan should be used to ensure good visualisation of any pathology in the pancreas.

Although there was no direct evidence on FDG-PET/CT as a diagnostic test for pancreatic solid lesions, the committee believed that the evidence regarding its use in the diagnosis of pancreatic cancer in people with jaundice (see section 5.1) and of people without jaundice but with pancreatic abnormalities such as cysts (as described in Ghaneh et al. 2018 – see section 5.3) merited its wider use in the diagnosis of people with solid lesions. As such, the committee believed that FDG-PET/CT will add significant additional information, particularly with respect to detecting metastatic disease if the diagnosis is unclear after the intial CT scan. The committee noted that EUS with tissue sampling had both high sensitivity and specificity whereas FDG-PET/CT had high sensitivity but lower specificity. The committee decided that the non-invasive nature of FDG-PET/CT, the low false negative rate and the additional information related to metastic disease that it can provide, would put FDG-PET/CT alongside EUS with tissue sampling as the next step if further diagnostic information is required after the CT scan. For these reasons even though there was a lack of direct evidence the committee decided, based on consensus, to make a strong recommendation that a FDG-PET/CT scan should be conducted and / or EUS (with tissue sampling) if the diagnosis is still unclear after CT.

The committee noted that EUS-guided tissue sampling can provide cytology or histology, which a CT scan is unable to do. Based on their knowledge and experience, the committee agreed that having cytology or histology would help to resolve diagnostic uncertainty, facilitate oncological management and is needed to enrol people in clinical trials. Therefore, based on the evidence and their knowledge, the committee agreed to recommend EUS-guided tissue sampling for those people whose CT scan was inconclusive. They were unable to specify whether FNA or FNB should be used for the tissue sampling as the evidence did not support recommending 1 method over another. The committee considered that the potential benefits of the recommendations made would be more accurate diagnosis of pancreatic cancer in people with a solid lesion. The potential harms of the recommendations were the potential for complications associated with EUS-guided tissue sampling. However, the committee agreed that the benefits outweighed the harms as tissue sampling was only recommended for a sub-set of the people being investigated.

5.2.8.4. Consideration of economic benefits and harms

The committee noted that whilst no relevant published economic evaluations were identified for this topic, diagnosis (including patients with jaundice) formed part of the diagnosis and staging pathway for the cost utility analysis in a health technology assessment (HTA) by Ghaneh et al. (2018) identified for staging and discussed in detail in section 7.5.1.

The HTA highlighted that including FDG-PET/CT as part of the diagnostic and staging work up of patients with suspected pancreatic cancer was very likely cost saving and health improving. It was acknowledged that the HTA did not look at the cost effectiveness of the addition of FDG-PET/CT in a sub-group of patients without jaundice but with pancreatic abnormalities although this group would be a large component of study cohort considered. It was noted that the definition of pancreatic abnormality for the inclusion criteria in the HTA study (focal lesion in the pancreas/bulky pancreas/dilated pancreatic duct) was more restrictive than the definition used for this question although it would account for the majority of such abnormalities and the committee were confident that the evidence from this study could be extrapolated since it also included people with pancreatic cysts.

The recommendations related to the topic in this section, as well as those for diagnosis of people with suspected pancreatic cancer with jaundice and for staging almost identically match the diagnosis and staging pathway used as the intervention in the HTA’s cost utility study. The committee therefore considered the reasons discussed in section 7.8.4. applied to the two diagnostic recommendations as well. The committee were therefore confident this recommendation was cost effective and very likely cost saving and health improving.

As for diagnosis for patients with jaundice (see section 5.1.7) this recommendation in favour of FDG-PET/CT impacts upon a large proportion of the population considered for this guideline. There will be an initial increase in resource use through increased imaging with more expensive FDG-PET/CT, but this is likely to be recouped within one year.

5.2.9. References

  • Bang JY, Hebert-Magee S, Trevino J et al. (2012) Randomized trial comparing the 22-gauge aspiration and 22-gauge biopsy needles for EUS-guided sampling of solid pancreatic mass lesions. Gastrointestinal Endoscopy 76(2): 321–327 [PMC free article: PMC4148209] [PubMed: 22658389]
  • Bournet B, Selves J, Grand D et al. (2015) Endoscopic Ultrasound–guided Fine-Needle Aspiration Biopsy Coupled with a KRAS Mutation Assay Using Allelic Discrimination Improves the Diagnosis of Pancreatic Cancer. Journal of Clinical Gastroenterology 49(1): 50–56 [PubMed: 24798941]
  • Bournet B, Souque A, Senesse P et al. (2009) Endoscopic ultrasound-guided fine-needle aspiration biopsy coupled with KRAS mutation assay to distinguish pancreatic cancer from pseudotumoral chronic pancreatitis. Endoscopy 41(06): 552–557 [PubMed: 19533561]
  • Fabbri C, Polifemo AM, Luigiano C et al. (2011) Endoscopic ultrasound-guided fine needle aspiration with 22-and 25-gauge needles in solid pancreatic masses: a prospective comparative study with randomisation of needle sequence. Digestive and Liver Disease 43(8): 647–652 [PubMed: 21592873]
  • Fritscher-Ravens A, Brand L, Knöfel WT et al. (2002) Comparison of endoscopic ultrasound-guided fine needle aspiration for focal pancreatic lesions in patients with normal parenchyma and chronic pancreatitis. The American Journal of Gastroenterology, 97(11): 2768–2775 [PubMed: 12425546]
  • Ghaneh P, Hanson R, Titman A et al. (2018) PET-PANC: multicentre prospective diagnostic accuracy and health economic analysis study of the impact of combined modality 18fluorine-2-fluoro-2-deoxy-d-glucose positron emission tomography with computed tomography scanning in the diagnosis and management of pancreatic cancer. Health Technology Assessment 22(7) [PMC free article: PMC5817411] [PubMed: 29402376]
  • Harewood GC and Wiersema MJ (2002) Endosonography-guided fine needle aspiration biopsy in the evaluation of pancreatic masses. The American Journal of Gastroenterology 97(6): 1386–1391 [PubMed: 12094855]
  • Hikichi T, Irisawa A, Bhutani MS et al. (2009) Endoscopic ultrasound-guided fine-needle aspiration of solid pancreatic masses with rapid on-site cytological evaluation by endosonographers without attendance of cytopathologists. Journal of Gastroenterology 44(4): 322–328 [PubMed: 19274426]
  • Iglesias-Garcia J, Dominguez-Munoz E, Lozano-Leon A et al. (2007) Impact of endoscopic ultrasound-guided fine needle biopsy for diagnosis of pancreatic masses. World Journal of Gastroenterology 13(2): 289 [PMC free article: PMC4065960] [PubMed: 17226911]
  • Kliment M, Urban O, Cegan M et al. (2010) Endoscopic ultrasound-guided fine needle aspiration of pancreatic masses: the utility and impact on management of patients. Scandinavian Journal of Gastroenterology 45(11): 1372–1379 [PubMed: 20626304]
  • Krishna NB, LaBundy JL, Saripalli S et al. (2009) Diagnostic value of EUS-FNA in patients suspected of having pancreatic cancer with a focal lesion on CT scan/MRI but without obstructive jaundice. Pancreas 38(6): 625–630 [PubMed: 19506529]
  • Lee YN, Moon JH, Kim HK et al. (2014) Core biopsy needle versus standard aspiration needle for endoscopic ultrasound-guided sampling of solid pancreatic masses: a randomized parallel-group study. Endoscopy 46(12): 1056–1062 [PubMed: 25098611]
  • Mishra G, Zhao Y, Sweeney J et al. (2006) Determination of qualitative telomerase activity as an adjunct to the diagnosis of pancreatic adenocarcinoma by EUS-guided fine-needle aspiration. Gastrointestinal Endoscopy 63(4): 648–654 [PubMed: 16564867]
  • Ramesh J, Bang JY, Hebert-Magee S et al. (2015) Randomized trial comparing the flexible 19G and 25G needles for endoscopic ultrasound-guided fine needle aspiration of solid pancreatic mass lesions. Pancreas 44(1): 128–133 [PMC free article: PMC4272223] [PubMed: 25232713]
  • Seicean A, Gheorghiu M, Zaharia T et al. (2016) Performance of the Standard 22G Needle for Endoscopic Ultrasound-guided Tissue Core Biopsy in Pancreatic Cancer. Journal of Gastrointestinal Liver Disease 25(2): 213–218 [PubMed: 27308653]
  • Strand DS, Jeffus SK, Sauer BG et al. (2014) EUS – guided 22 – gauge fine – needle aspiration versus core biopsy needle in the evaluation of solid pancreatic neoplasms. Diagnostic Cytopathology 42(9): 751–758 [PubMed: 24550162]
  • Tamm EP, Loyer EM, Faria SC et al. (2007) Retrospective analysis of dual-phase MDCT and follow-up EUS/EUS-FNA in the diagnosis of pancreatic cancer. Abdominal Imaging 32(5): 660–667 [PubMed: 17712589]
  • Touchefeu Y, Le Rhun M, Coron E et al. (2009) Endoscopic ultrasound – guided fine – needle aspiration for the diagnosis of solid pancreatic masses: the impact on patient – management strategy. Alimentary Pharmacology & Therapeutics 30(10): 1070–1077 [PubMed: 19735232]
  • Wakatsuki T, Irisawa A, Bhutani MS et al. (2005) Comparative study of diagnostic value of cytologic sampling by endoscopic ultrasonography – guided fine – needle aspiration and that by endoscopic retrograde pancreatography for the management of pancreatic mass without biliary stricture. Journal of Gastroenterology and Hepatology 20(11): 1707–1711 [PubMed: 16246190]
  • Wittmann J, Kocjan G, Sgouros SN et al. (2006) Endoscopic ultrasound-guided tissue sampling by combined fine needle aspiration and trucut needle biopsy: a prospective study. Cytopathology 17(1): 27–33 [PubMed: 16417562]
  • Yang RY, Ng D, Jaskolka JD et al. (2015) Evaluation of percutaneous ultrasound-guided biopsies of solid mass lesions of the pancreas: a center’s 10-year experience. Clinical Imaging 39(1): 62–65 [PubMed: 25043532]
  • Yusuf TE, Ho S, Pavey DA et al. (2009) Retrospective analysis of the utility of endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) in pancreatic masses, using a 22-gauge or 25-gauge needle system: a multicenter experience. Endoscopy 41(05): 445–448 [PubMed: 19418399]

5.3. Pancreatic Cysts

Review question: In adults with a pancreatic cyst, what is the diagnostic pathway to identify the cyst(s) at high risk of pancreatic malignancy?

5.3.1. Introduction

The diagnosis of pancreatic cysts continues to increase in frequency as more people undergo cross sectional imaging.

The morphological identification of a cyst is straightforward on both MRI and CT but the identification of the exact nature of the cystic lesion continues to present diagnostic difficulty.

Three broad groups of cystic lesions can be identified; definitely malignant, definitely benign and indeterminate. There are features on imaging that suggest a cyst is suspicious in nature, but often these are not definitive.

The presence of mucin within the cyst and the measurement of markers such as Carcinoembryonic antigen (CEA) and amylase can help determine whether a lesion is benign or pre-malignant, and the role of cytology and histology is important.

Several diagnostic pathways have been suggested within the literature but there remains inconsistency within the UK as to the most effective method for diagnosis.

Guidance is needed on the most effective diagnostic pathway to identify cysts at high risk of malignancy in people with pancreatic cysts.

5.3.1.1. Review protocol summary

The review protocol summary used for this question can be found in Table 34. Full details of the review protocol can be found in Appendix C.

Table 34. Clinical review protocol summary for the review of most effective diagnostic pathway to identify the cyst(s) at high risk of pancreatic malignancy.

Table 34

Clinical review protocol summary for the review of most effective diagnostic pathway to identify the cyst(s) at high risk of pancreatic malignancy.

5.3.2. Description of Clinical Evidence

Thirty-five publications were included in this review: 2 of these were systematic reviews (Cao et al. 2016; Zhu et al. 2017), 6 were prospective cohort studies (Brugge et al. 2004; Cizginer et al. 2011; Frossard et al. 2003; Ghaneh et al. 2007; Pitman et al. 2013; Sperti et al. 2005), and 27 of them were retrospective cohort studies (Ardengh et al. 2007; Gaddam et al. 2015; Gerke et al. 2006; Hirono et al. 2012; Jang et al. 2014; Jin et al. 2015; Kamata et al. 2016; Kim et al. 2012; Kim et al. 2015; Lee et al. 2001; Linder et al. 2006; Moris et al. 2016; Nagashio et al. 2014; Nara et al. 2009; Oh et al. 2014; Oppong et al. 2015; Othman et al. 2012; Pais et al. 2007; Park et al. 2011; Pitman et al. 2010; Smith et al. 2016; Song et al. 2007; Sperti et al. 2001; Takanami et al. 2011; Talar-Wojnarowska et al. 2013; Wu et al. 2007; Zhang et al. 2010). A summary of the included studies is presented in Table 36.

Fourteen studies examined the diagnostic accuracy of cyst fluid analysis, cytology or imaging for distinguishing between mucinous cystic neoplasms (MCNs; including IPMNs) and non-mucinous cystic neoplasms (NMCNs) of the pancreas (Brugge et al. 2004; Cizginer et al. 2011; Frossard et al. 2003; Gaddam et al. 2015; Jin et al. 2015; Linder et al. 2006; Moris et al. 2016; Nagashio et al. 2014; Oh et al. 2014; Oppong et al. 2015; Park et al. 2011; Pitman et al. 2010; Song et al. 2007; Zhang et al. 2010).

Twenty studies examined the diagnostic accuracy of cyst fluid analysis, cytology or imaging for distinguishing between benign and potentially malignant or malignant pancreatic cystic lesions (PCLs) (Ardengh et al. 2007; Cao et al. 2016; Gerke et al. 2006; Ghaneh et al. 2018; Hirono et al. 2012; Jang et al. 2014; Kamata et al. 2016; Kim et al. 2012; Kim et al. 2015; Lee et al. 2011; Nara et al. 2009; Othman et al. 2012; Pais et al. 2007; Pitman et al. 2013; Smith et al. 2016; Sperti et al. 2001, Sperti et al. 2005; Takanami et al. 2011; Talar-Wojnarowska et al. 2013; Wu et al. 2007).

One study (Park et al. 2011) examined the diagnostic accuracy of cyst fluid analysis, cytology or imaging for distinguishing between both (i) MCNs and NMCNs and (ii) benign and potentially malignant PCLs.

One of the systematic reviews (Cao et al. 2016) aimed to evaluate the diagnostic value of serum CA 19-9 in identifying malignant PCLs and included 13 studies (n=1437). The other systematic review (Zhu et al. 2017) evaluated the morbidity and mortality associated with EUS-FNA for the diagnosis of PCLs, and included 40 studies (n=5147). Both systematic reviews were assessed as being of high methodological quality, but included very low to moderate quality evidence. See Table 36 for more details of the included studies.

Positive and likelihood ratios were calculated, where appropriate, from the raw diagnostic test accuracy data or the estimated sensitivity and specificity of the studies to enable evaluation of the relevant tests. The QUADAS-2 tool was used for assessing risk of bias and indirectness of included studies.

Further information about the search strategy can be found in Appendix D. See study selection flow chart in Appendix E, single and multiple test ROC curves and forest plots in Appendix H, summary of QUADAS-2 study quality evaluations in Appendix J, study evidence tables in Appendix F and list of excluded studies in Appendix G.

5.3.2.1. CEA

5.3.2.1.1. Cystic fluid CEA

Thirteen studies (n=1542) examined the diagnostic accuracy of cyst fluid CEA: 2 of these were prospective cohort studies (Brugge et al. 2004; Cizginer et al. 2011), whilst the remaining 11 were retrospective cohort studies. The median number of patients was 112 (range 52-226).

Nine studies focused on distinguishing between MCNs and NMCNs (Brugge et al. 2004; Cizginer et al. 2011; Gaddam et al. 2015; Jin et al. 2015; Linder et al. 2006; Moris et al. 2016; Nagashio et al. 2014; Oppong et al. 2015; Oh et al. 2014). One study examined the diagnostic accuracy of CEA for distinguishing between both types of cystic lesions (Park et al. 2011). The cut-off value of cystic fluid CEA used to differentiate pancreatic MCNs and NMCNs ranged from 5 to 6000 ng/ml, and were categorised as detailed in Table 35:

Table 35. Studies on cystic fluid CEA by cut-off level.

Table 35

Studies on cystic fluid CEA by cut-off level.

Three studies evaluated the diagnostic accuracy of cyst fluid CEA for distinguishing between benign from potentially malignant and malignant PCLs (Hirono et al. 2012; Othman et al. 2012; Talar-Wojnarowska et al. 2013). The cut-off value of cystic fluid CEA used to differentiate benign from malign cysts ranged from 30 to 6000 ng/ml, and were categorised as follow:

5.3.2.1.2. Serum CEA

One retrospective study (n= 85) conducted in Taiwan evaluated serum levels of CEA for the differential diagnosis of pancreatic cystadenoma (benign PLC) or cystadenocarcinoma (malign PLC) (Wu et al. 2007).

5.3.2.2. CA 19-9

5.3.2.2.1. Cystic fluid CA 19-9

One meta-analysis (n=1437; Cao et al. 2016) of 13 observational studies (Fritz et al. 2011; Goh et al. 2008; Grobmyer et al. 2009; Hirono et al. 2012; Hwang et al. 2011; Ingkakul et al. 2010; Jones et al. 2009; Kitagawa et al. 2003; Ohtsuka et al. 2012; Sadakari et al. 2010; Shin et al. 2010; Sperti et al. 2007; and Xu et al. 2011) and 1 additional retrospective study (n=52; Talar-Wojnarowska et al. 2013) examined the diagnostic accuracy of CA 19-9 for distinguishing between benign and potentially malignant and malignant PCLs. The cut-off levels ranged from 35 to 45 ng/ml.

5.3.2.2.2. Serum CA 19-9

One study (n=85) conducted in Taiwan evaluated serum levels of CA 19-9 (Wu et al. 2007) for the differential diagnosis of pancreatic cystadenoma (benign PLC) or cystadenocarcinoma (malign PLC) (Wu, Yan et al. 2007).

5.3.2.3. Cytology: EUS-FNA

Ten studies (n=1164), 4 prospective and 6 retrospective cohort, examined the diagnostic accuracy of EUS-FNA cytology (Ardengh et al. 2007; Brugge et al. 2004; Cizginer et al. 2011; Frossard et al. 2003; Oppong et al. 2015; Pais et al. 2007; Pitman et al. 2010; Pitman et al. 2013; Smith et al. 2016; Zhang et al. 2010). Six of the studies evaluated the diagnostic accuracy of EUS-FNA based cytology for distinguishing between pancreatic MCNs and NMCNs (Brugge et al. 2004; Cizginer et al. 2011; Frossard et al. 2003; Oppong et al. 2015; Pitman et al. 2010; Zhang et al. 2010), whilst the remaining studies focused on distinguishing benign from potentially malignant or malignant PCLs (Ardengh et al. 2007; Pais et al. 2007; Pitman et al. 2013; Smith et al. 2016).

5.3.2.4. Imaging: CT

Seven studies (n=936), 2 prospective and 5 retrospective cohort, examined the diagnostic accuracy of CT (Gerke et al. 2006; Ghaneh et al. 2018; Lee et al. 2011; Nara et al. 2009; Song et al. 2007; Sperti et al. 2001; Sperti et al. 2005). Six of the studies focused on distinguishing between benign from potentially malignant and malignant PCLs (Gerke et al. 2006; Ghaneh et al. 2018; Lee et al. 2011; Nara et al. 2009; Sperti et al. 2001; Sperti et al. 2005).

5.3.2.5. Imaging: EUS

Seven studies (n=670), 3 prospective and 4 retrospective cohort, examined the diagnostic accuracy of EUS for the morphological evaluation of suspected pancreatic cystic neoplasms (Brugge et al. 2004; Cizginer et al. 2011; Frossard et al. 2003; Gerke et al. 2006; Kamata et al. 2016; Kim et al. 2012; Oppong et al. 2015). Three of the studies evaluated the accuracy of EUS for distinguishing between pancreatic MCNs and NMCNs (Gerke et al. 2006; Kamata et al. 2016; Kim et al. 2012); 4 studies focused on distinguishing between benign from potentially malignant and malignant PCLs (Brugge et al. 2004; Cizginer et al. 2011; Frossard et al. 2003; Oppong et al. 2015); and 3 studies evaluated the accuracy of EUS.

5.3.2.6. Imaging: EUS-FNA

One retrospective cohort study (n=119) examined the diagnostic accuracy of EUS-FNA for distinguishing between pancreatic MCNs and NMCNs (Oppong et al. 2015).

5.3.2.7. Imaging: FDG-PET/CT

Four studies (n=715), 2 prospective and 2 retrospective, examined the diagnostic accuracy of 18-fluorodeoxyglucose PET in distinguishing benign from malignant cystic lesions of the pancreas (Ghaneh et al. 2018; Sperti et al. 2001; Sperti et al. 2005; Takanami et al. 2011). The most recent study (Ghaneh et al. 2018), known as PET-PANC, was a multicentre UK study and used a standardised protocol to examine whether the addition of FDG-PET/CT to MDCT provides tangible diagnostic and staging benefits.

5.3.2.8. Imaging: MRI

Five retrospective cohort studies (n=324) examined the diagnostic accuracy of MRI: 4 of these (n=271) examined the diagnostic accuracy of MRI for distinguishing benign from malignant PCLs (Jang et al. 2014; Kim et al. 2012; Kim et al. 2015; and Lee et al. 2011), whilst 1 of these examined the accuracy of MRI in the differentiation of IPMNs from other pancreatic cystic masses (n=53; Song et al. 2007).

5.3.3. Summary of included studies

A summary of the studies that were included in this review is presented in Table 36

Table 36. Summary of included studies.

Table 36

Summary of included studies.

5.3.4. Clinical evidence profile

The clinical evidence profiles for this review are presented in Table 39 to Table 54

5.3.4.1. Cystic fluid or serum CEA

5.3.4.1.1. Cystic fluid CEA
Table 37. Summary of clinical evidence for meta-analyses of cystic fluid CEA to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 37

Summary of clinical evidence for meta-analyses of cystic fluid CEA to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 38. Summary of clinical evidence for other studies on cystic fluid CEA at various cut-offs to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 38

Summary of clinical evidence for other studies on cystic fluid CEA at various cut-offs to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 39. Summary of clinical evidence for studies on cystic fluid CEA to distinguish between (potentially) malignant and benign pancreatic cystic lesions.

Table 39

Summary of clinical evidence for studies on cystic fluid CEA to distinguish between (potentially) malignant and benign pancreatic cystic lesions.

5.3.4.1.2. Serum CEA
Table 40. Summary of clinical evidence for studies on serum CEA to distinguish between benign and (potentially) malignant pancreatic cystic lesions.

Table 40

Summary of clinical evidence for studies on serum CEA to distinguish between benign and (potentially) malignant pancreatic cystic lesions.

5.3.4.2. Cystic fluid or serum CA 19-9

5.3.4.2.1. Cystic fluid CA 19-9
Table 41. Summary of clinical evidence for meta-analysis of cystic fluid CA 19-9 to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 41

Summary of clinical evidence for meta-analysis of cystic fluid CA 19-9 to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

5.3.4.2.2. Serum CA 19-9
Table 42. Summary of clinical evidence for studies on serum CA 19-9 to distinguish between malignant and benign pancreatic cystic lesions.

Table 42

Summary of clinical evidence for studies on serum CA 19-9 to distinguish between malignant and benign pancreatic cystic lesions.

5.3.4.3. Cytology: EUS-FNA

Table 43. Summary of clinical evidence for meta-analysis of EUS-FNA cytology to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 43

Summary of clinical evidence for meta-analysis of EUS-FNA cytology to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 44. Summary of clinical evidence for meta-analysis of EUS-FNA cytology to distinguish between malignant and benign pancreatic cystic lesions.

Table 44

Summary of clinical evidence for meta-analysis of EUS-FNA cytology to distinguish between malignant and benign pancreatic cystic lesions.

5.3.4.4. Imaging: CT

Table 45. Summary of clinical evidence for studies on computed tomography to distinguish between mucinous cystic and non- mucinous cystic neoplasms of the pancreas.

Table 45

Summary of clinical evidence for studies on computed tomography to distinguish between mucinous cystic and non- mucinous cystic neoplasms of the pancreas.

Table 46. Summary of clinical evidence for meta-analysis of computed tomography to distinguish between malignant and benign pancreatic cystic lesions.

Table 46

Summary of clinical evidence for meta-analysis of computed tomography to distinguish between malignant and benign pancreatic cystic lesions.

5.3.4.5. Imaging: EUS

Table 47. Summary of clinical evidence for meta-analysis of EUS to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 47

Summary of clinical evidence for meta-analysis of EUS to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 48. Summary of clinical evidence for studies on EUS to distinguish between malignant and benign pancreatic cystic lesions.

Table 48

Summary of clinical evidence for studies on EUS to distinguish between malignant and benign pancreatic cystic lesions.

5.3.4.6. Imaging: EUS-FNA

Table 49. Summary of clinical evidence for studies on EUS-FNA to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 49

Summary of clinical evidence for studies on EUS-FNA to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

5.3.4.7. Imaging: FDG-PET/CT

Table 50. Summary of clinical evidence for studies on FDG-PET/CT to distinguish between (potentially) malignant and benign pancreatic cystic lesions.

Table 50

Summary of clinical evidence for studies on FDG-PET/CT to distinguish between (potentially) malignant and benign pancreatic cystic lesions.

5.3.4.8. Imaging: MRI

Table 51. Summary of clinical evidence for meta-analysis of MRI to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 51

Summary of clinical evidence for meta-analysis of MRI to distinguish between mucinous cystic and non-mucinous cystic neoplasms of the pancreas.

Table 52. Summary of clinical evidence for studies on MRI to distinguish between (potentially) malignant and benign pancreatic cystic lesions.

Table 52

Summary of clinical evidence for studies on MRI to distinguish between (potentially) malignant and benign pancreatic cystic lesions.

5.3.5. Economic evidence

A literature review of published cost effectiveness analyses did not identify any relevant studies for this topic. Although there were potential implications for resource use associated with making recommendations in this area, other topics in the guideline were agreed as a higher economic priority. Consequently, bespoke economic modelling was not done for this topic.

5.3.6. Evidence statements

5.3.6.1. Carcinoembryonic antigen (CEA) tests

5.3.6.1.1. Cystic fluid CEA
Mucinous cystic neoplasms versus non-mucinous cystic neoplasms of the pancreas
Diagnostic accuracy

Moderate quality evidence from a meta-analysis of 4 cohort studies (1 prospective and 3 retrospective) (n=401) found that cystic fluid CEA with a cut-off level of 192 ng/ml had a low sensitivity of 0.58 (95% CI, 0.49-0.67) and a moderate specificity of 0.87 (95% CI, 0.74-0.94) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 4.33 (95% CI, 2.27-8.26) suggests that a positive result for a mucinous cystic neoplasm is not particularly useful for ruling it in, though there is uncertainty in the estimate. The negative likelihood ratio of 0.48 (95% CI, 0.39-0.59) suggests that a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it in or ruling it out.

Low quality evidence from a meta-analysis of 5 retrospective cohort studies (n=434) found that cystic fluid CEA with a cut-off level of between 30 and 70 ng/ml had a moderate sensitivity of 0.88 (95% CI, 0.82-0.92) and moderate specificity of 0.82 (95% CI, 0.72–0.89) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 4.83 (95% CI, 3.08-7.58) suggests that a positive result for a mucinous cystic neoplasm is not particularly useful in ruling it in, though there is uncertainty in the estimates. The negative likelihood ratio of 0.15 (0.1-0.23) suggests that a negative result for a mucinous cystic neoplasm is moderately useful for ruling it out, though there is uncertainty in the estimates.

Low quality evidence from 1 retrospective cohort study (n=226) found that cystic fluid CEA with a cut-off level of 5 ng/ml had a high sensitivity of 0.94 (95% CI, 0.89-0.97) and a low specificity of 0.42 (95% CI, 0.31-0.54) for distinguishing between mucinous and non mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 1.62 (95% CI, 1.33-1.98) suggests that a positive result for a mucinous cystic neoplasm is not particularly useful for ruling it in. The negative likelihood ratio of 0.4 (95% CI, 0.07-0.28) suggests that neither a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it out, though there is substantial uncertainty in the estimate.

Very low quality evidence from 1 retrospective cohort study (n=78) found that cystic fluid CEA with a cut-off level of 7 ng/ml had a high sensitivity of 0.94 (95% CI, 0.83-0.99) and a moderate specificity of 0.75 (95% CI, 0.55-0.89) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 3.76 (95% CI, 1.97-7.17) suggests that a positive result for a mucinous cystic neoplasm is not particularly useful in ruling it in, though there is uncertainty in the estimate. The negative likelihood ratio of 0.08 (95% CI, 0.03-0.24) suggests that a negative result for a mucinous cystic neoplasm is very useful for ruling it out, though there is substantial uncertainty in the estimate.

Very low quality evidence from 1 retrospective cohort study (n=226) found that cystic fluid CEA with a cut-off level of 105 ng/ml had a moderate sensitivity of 0.7 (95% CI, 0.62-0.77) and a low specificity of 0.63 (95% CI, 0.51-0.74) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 1.9 (95% CI, 1.39-2.6) and negative likelihood ratio of 0.48 (95% CI, 0.35-0.64) suggests that neither a positive or negative result for a mucinous cystic neoplasm is particularly useful for ruling it in or ruling it out.

Very low quality evidence from 2 cohort studies (1 prospective and 1 retrospective) (n=436) found that cystic fluid CEA with a cut-off level of 110 ng/ml had a low to moderate sensitivity ranging from 0.62 to 0.81 and a high specificity ranging from 0.93 to 0.98 for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratios were 8.68 (95% CI, 2.24-33.58) to 35.6 (5.12-247.66) suggesting that a positive result for a mucinous cystic neoplasm is either moderately useful or very useful for ruling it in, though there is substantial uncertainty in the estimates. The negative likelihood ratios were 0.2 (95% CI, 0.13-0.29) and 0.41 (95% CI, 0.28-0.59) suggesting that a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it out, though there is uncertainty in the estimates.

Low quality evidence from 1 retrospective cohort study (n=180) found that cystic fluid CEA with a cut-off level of 129 ng/ml had a moderate sensitivity of 0.77 (95% CI, 0.7-0.84) and a moderate specificity of 0.83 (95% CI, 0.66-0.93) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 4.51 (95% CI, 2.16-9.38) suggests that a positive result for a mucinous cystic neoplasm is not particularly useful for ruling it out, though there is uncertainty in the estimate. The negative likelihood ratio of 0.27 (95% CI, 0.2-0.38) suggests that a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it out.

Moderate quality evidence from 1 retrospective cohort study (n=124) found that cystic fluid CEA with a cut-off level of 200 ng/ml had a low sensitivity of 0.6 (95% CI, 0.49-0.71) and a high specificity of 0.93 (95% CI, 0.81-0.99) for distinguishing between mucinous and non mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 8.67 (95% CI, 2.87-26.19) suggests that a positive result for a mucinous cystic neoplasm is moderately useful for ruling it in, though there is substantial uncertainty in the estimate. The negative likelihood ratio of 0.42 (95% CI, 0.32-0.56) suggests that a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it out.

Very low quality evidence from 1 retrospective cohort study (n=71) found that cystic fluid CEA with a cut-off level of 300 ng/ml had a low sensitivity of 0.41 (95% CI, 0.3-0.53) and a moderate specificity of 0.89 (95% CI, 0.52-1.0) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 3.86 (95% CI, 0.6-24.92) suggests that a positive result for a mucinous cystic neoplasm is not particularly useful for ruling it in, though there is substantial uncertainty in the estimate. The negative likelihood ratio of 0.64 (95% CI, 0.48-0.87) suggests that a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it out.

Low quality evidence from 3 retrospective cohort studies (n=436) found that cystic fluid CEA with a cut-off level of 800 ng/ml had a low sensitivity ranging from 0.27 to 0.38 and a moderate to high specificity ranging from 0.86 to 0.95 for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratios were 2.3 (95% CI, 1.27-4.16), 2.45 (95% CI, 0.37-16.14) to 8.23 (95% CI, 2.07-32.75) suggesting that a positive result for a mucinous cystic neoplasm is either not particularly useful or moderately useful, though there is uncertainty in the estimates the negative likelihood ratios were 0.65 (95% CI, 0.57-0.78), 0.78 (95% CI, 0.67-0.9) to 0.82 (95% CI, 0.63-1.07) suggesting that a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it out.

Moderate quality evidence from 1 retrospective cohort study (n=71) found that cystic fluid CEA with a cut-off level of 6000 ng/ml had a moderate sensitivity of 0.86 (95% CI, 0.7-0.95) and a high specificity of 1.0 (0.9-1.0) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 62.69 (95% CI, 3.98-987.16) suggests that a positive result for a mucinous cystic neoplasm is very useful for ruling it in, though there is substantial uncertainty in the estimate. The negative likelihood ratio of 0.14 (95% CI, 0.06-0.32) suggests that a negative result for a mucinous cystic neoplasm is moderately useful for ruling it out, though there is substantial uncertainty in the estimate.

Adverse events

No evidence was identified to inform this outcome.

Malignant versus benign pancreatic cystic lesions
Diagnostic accuracy

Very low quality evidence from 1 retrospective cohort study (n=134) found that cystic fluid CEA with a cut-off level of 30 ng/ml had a high sensitivity of 0.95 (95% CI, 0.85-0.99) and a moderate specificity of 0.85 (95% CI, 0.75-0.92) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 6.15 (95% CI, 3.64-10.39) suggests that a positive result for malignancy is moderately useful for ruling it in, though there is substantial uncertainty in the estimate. The negative likelihood ratio of 0.06 (95% CI, 0.02-0.19) suggests that a negative result for malignancy is very useful for ruling it out, though there is uncertainty in the estimate.

Low quality evidence from 1 retrospective cohort study (n=52) found that cystic fluid CEA with a cut-off level of 45 ng/ml had a high sensitivity of 0.94 (95% CI, 0.7-1.0) and a low specificity of 0.64 (95% CI, 0.46-0.79) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 2.6 (95% CI, 1.65-4.08) suggests that positive result for malignancy is not particularly useful for ruling it in, whilst the negative likelihood ratio of 0.1 (95% CI, 0.01-0.66) suggests that a negative result for malignancy is moderately useful in ruling it out, though there is substantial uncertainty in the estimate.

Low quality evidence from 1 retrospective cohort study (n=63) found that cystic fluid CEA with a cut-off level of 6000 ng/ml had a low sensitivity of 0.31 (95% CI, 0.11-0.59) and moderate specificity of 0.85 (95% CI, 0.72-0.94) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 2.1 (95% CI, 0.77-5.69) suggests that a positive result for malignancy is not particularly useful for ruling it in, though threre is uncertainty in the estimate. The negative likelihood ratio of 0.81 (95% CI, 0.57-1.15) suggests that a negative result for malignancy is not particularly useful for ruling it out.

Adverse events

No evidence was identified to inform this outcome.

5.3.6.1.2. Serum CEA
Diagnostic accuracy

Low quality evidence from 1 retrospective study (n= 85), which did not specify the cut-off level, found that serum CEA had a low sensitivity of 0.35 (95% CI, 0.22-0.51) and moderate specificity of 0.84 (95% CI, 0.68-0.94) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 2.18 (95% CI, 0.96-4.99) and negative likelihood ratio of 0.77 (95% CI, 0.6-0.99) suggest that neither a positive or negative result for malignancy is particularly useful for ruling it and ruling it out.

Adverse events

No evidence was identified to inform this outcome.

5.3.6.2. Cancer antigen 19-9 (CA 19-9) test

5.3.6.2.1. Cystic fluid CA 19-9
Diagnostic accuracy

Moderate quality evidence from a meta-analysis of 14 studies (n=1489) found that cystic fluid CA 19-9 at a cut-off of between 35 and 45 ng/ml had a low sensitivity of 0.5 (95% CI, 0.37-0.63) and moderate specificity of 0.87 (95% CI, 0.84-0.9) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 3.92 (95% CI, 3.16-4.87) and negative likelihood ratio of 0.58 (95% CI, 0.46-0.73) suggest that neither a positive or negative result for a mucinous cystic neoplasm is particularly useful for ruling it in and ruling it out.

Adverse events

No evidence was identified to inform this outcome.

5.3.6.2.2. Serum CA 19-9
Diagnostic accuracy

Low quality evidence from 1 retrospective study (n= 85), which did not specify the cut-off level, found that serum CA 19-9 had a low sensitivity of 0.58 (95% CI, 0.43-0.72) and moderate specificity of 0.86 (95% CI, 0.71-0.95) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 4.32 (95% CI, 1.85-10.09) suggest that a positive result for malignancy is not particularly useful for ruling it in, though there is substantial uncertainty in the estimate. The negative likelihood ratio of 0.48 (95% CI, 0.34-0.69) suggest that a negative result for malignancy is not particularly useful for ruling it out.

Adverse events

No evidence was identified to inform this outcome.

5.3.6.3. Cytology: EUS-FNA

Mucinous cystic neoplasms versus non-mucinous cystic neoplasms of the pancreas
Diagnostic accuracy

Very low quality evidence from a meta-analysis of 6 cohort studies (3 prospective and 3 retrospective) (n=639) found EUS-FNA-based cytology had a low sensitivity of 0.55 (95% CI, 0.27-0.8) and high specificity of 0.94 (95% CI, 0.86-0.97) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 8.52 (95% CI, 3.41-21.31) suggests that a positive result for a mucinous cystic neoplasm is moderately useful for ruling it in, though there is substantial uncertainty in the estimate, the negative likelihood ratio of 0.48 (95% CI, 0.25-0.91) suggests that a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it out.

Adverse events

No evidence was identified to inform this outcome.

Malignant versus benign pancreatic cystic lesions
Diagnostic accuracy

Low quality evidence from a meta-analysis of 4 cohort studies (1 prospective and 3 retrospective) (n=454) found that EUS-FNA-based cytology had a low sensitivity of 0.7 (95% CI, 0.54-0.81) and a high specificity of 0.93 (95% CI, 0.88-0.96) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 9.67 (95% CI, 6.14-15.24) suggests that a positive result for a mucinous cystic neoplasm is moderately useful for ruling it in, though there is uncertainty in the estimate. The negative likelihood ratio of 0.33 (95% CI, 0.21-0.5) suggests that a negative result for malignancy is not particularly useful for ruling it out.

Adverse effects

High quality evidence from a meta-analysis of 40 studies (n=5124) found that EUS-FNA cytology is a safe procedure for diagnosis of pancreatic cystic lesions and is associated with a relatively low incidence of adverse events.

5.3.6.4. Imaging: CT

Mucinous cystic neoplasms versus non-mucinous cystic neoplasms of the pancreas
Diagnostic accuracy

Low quality evidence from 1 retrospective cohort study (n=53) found that CT had a moderate sensitivity of 0.81 (95% CI, 0.63-0.93) and a moderate specificity of 0.86 (95% CI, 0.78-0.93) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 5.96 (95% CI, 3.49-10.16) suggests that a positive result for a mucinous cystic neoplasm is moderately useful for ruling it in, though there is substantial uncertainty in the estimate. The negative likelihood ratio of 0.22 (95% CI, 0.11-0.46) suggests that a negative result for a mucinous cystic neoplasm is not particularly useful for ruling it out, though there is uncertainty in the estimate.

Adverse events

No evidence was identified to inform this outcome.

Malignant versus benign pancreatic cystic lesions
Diagnostic accuracy

Very low quality evidence from a meta-analysis of 6 cohort studies (2 prospective and 4 retrospective) (n=883) found that CT had a low sensitivity of 0.69 (95% CI, 0.60-0.78) and a high specificity of 0.91 (95% CI, 0.89-0.93) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 8.00 (95% CI, 6.17-10.37) suggests that a positive result for malignancy is moderately useful for ruling it in, though there is uncertainty in the estimate. The negative likelihood ratio of 0.34 (95% CI, 0.26-0.44) suggests that a negative result for malignancy is not particularly useful for ruling it out.

Adverse events

In 1 multicentre prospective cohort study (n=583) that examined the diagnostic test accuracy of CT, no adverse events related to the tests were reported.

5.3.6.5. Imaging: EUS

Diagnostic accuracy

Very low quality evidence from a meta-analysis of 4 cohort studies (1 prospective and 3 retrospective) (n=210) found that EUS had a low sensitivity of 0.67 (95% CI, 0.43-0.84) and low specificity of 0.65 (95% CI, 0.48-0.78) for distinguishing between mucinous and non mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 1.88 (95% CI, 1.18-3.0) and negative likelihood ratio of 0.52 (95% CI, 0.28-0.96) suggests that neither a positive or negative result for a mucinous cystic neoplasm is particularly useful for ruling it in or ruling it out.

Adverse events

No evidence was identified to inform this outcome.

Mucinous cystic neoplasms versus non-mucinous cystic neoplasms of the pancreas
Diagnostic accuracy

Very low quality evidence from a meta-analysis of 4 cohort studies (1 prospective and 3 retrospective) (n=210) found that EUS had a low sensitivity of 0.67 (95% CI, 0.43-0.84) and low specificity of 0.65 (95% CI, 0.48-0.78) for distinguishing between mucinous and non mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 1.88 (95% CI, 1.18-3.0) and negative likelihood ratio of 0.52 (95% CI, 0.28-0.96) suggests that neither a positive or negative result for a mucinous cystic neoplasm is particularly useful for ruling it in or ruling it out.

Adverse events

No evidence was identified to inform this outcome.

Malignant versus benign pancreatic cystic lesions
Diagnostic accuracy

Very low quality evidence from 3 retrospective cohort studies (n=187) found that EUS had a low to high sensitivity ranging from 0.71 to 0.97 and a low specificity ranging from 0.4 to 0.73 for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratios were 1.61 (95% CI, 1.24-2.09), 1.91 (95% CI, 1.17-3.11) and 3.65 (95% CI, 1.57-8.45) suggesting that a positive result for malignancy is not particularly useful for ruling it in. The negative likelihood ratios were 0.04 (95% CI, 0.01-0.27), 0.08 (95% CI, 0.01-0.59) and 0.46 (95% CI, 0.25-0.85) suggesting that a negative result for malignancy is either very useful or not particularly useful in ruling it out, though there is substantial uncertainty in the estimates.

Adverse events

No evidence was identified to inform this outcome.

5.3.6.6. Imaging: EUS-FNA

Diagnostic accuracy

Low quality evidence from 1 retrospective study (n=119) found that EUS-FNA had a moderate sensitivity of 0.76 (95% CI, 0.65-0.85) and a low specificity of 0.73 (95% CI, 0.56--0.85) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 2.76 (95% CI, 1.64- 4.64) and negative likelihood ratio of 0.33 (95% CI, 0.21-0.51) suggests that neither a positive or negative result for a mucinous cystic neoplasm is particularly useful for ruling it in or ruling it out.

Adverse events

No evidence was identified to inform this outcome.

Malignant versus benign pancreatic cystic lesions
Diagnostic accuracy

Low quality evidence from 1 retrospective study (n=119) found that EUS-FNA had a moderate sensitivity of 0.76 (95% CI, 0.65-0.85) and a low specificity of 0.73 (95% CI, 0.56-0.85) for distinguishing between mucinous and non-mucinous cystic neoplasms of the pancreas in adults with pancreatic cysts. The positive likelihood ratio of 2.76 (95% CI, 1.64-4.64) and negative likelihood ratio of 0.33 (95% CI, 0.21-0.51) suggests that neither a positive or negative result for a mucinous cystic neoplasm is particularly useful for ruling it in or ruling it out.

Adverse events

No evidence was identified to inform this outcome.

5.3.6.7. Imaging: FDG-PET/CT

Malignant versus benign pancreatic cystic lesions
Diagnostic accuracy

Very low quality evidence from 4 cohort studies (2 prospective and 2 retrospective) (n=672) found that 18-FDG FDG-PET/CT had a moderate sensitivity of 0.86 (95% CI, 0.71-0.94) and a high specificity of 0.96 (95% CI, 0.94-0.97) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 20.8 (95% CI, 13.6-30.0) suggests that a positive result for malignancy is very useful for ruling it in. The negative likelihood ratio of 0.16 (95% CI, 0.06-0.29) suggests that a negative result for malignancy is moderately useful for ruling it out, though there is substantial uncertainty in the estimates.

Adverse events

In 1 multicentre prospective cohort study (n=583) that examined the diagnostic test accuracy of CT, no adverse events related to the tests were reported.

5.3.6.8. Imaging: MRI

Mucinous cystic neoplasms versus non-mucinous cystic neoplasms of the pancreas
Diagnostic accuracy

Moderate quality evidence from 1 retrospective study (n=53) found that MRI had a high sensitivity of 0.97 (95% CI, 0.83-1.0) and a high specificity of 0.91 (95% CI, 0.71-0.99) for distinguishing between non-mucinous and mucinous neoplasms. The positive likelihood ratio of 10.65 (95% CI, 2.84-39.97) and negative likelihood ratio of 0.04 (95% CI 0.01-0.25, suggest that both a positive and negative result for a mucinous cystic neoplasm are very useful for ruling it in and ruling it out, though there is substantial uncertainty in the estimates.

Adverse events

No evidence was identified to inform this outcome.

Malignant versus benign pancreatic cystic lesions
Diagnostic accuracy

Low quality evidence from a meta-analysis of 4 retrospective cohort studies (n=271) found that MRI had a moderate sensitivity of 0.79 (95% CI, 0.64-0.89) and a moderate sensitivity of 0.84 (95% CI, 0.69-0.92) for detecting malignancy or potential malignancy of pancreatic cystic lesions in adults. The positive likelihood ratio of 4.81 (95% CI, 2.54-9.08) and negative likelihood ratio of 0.25 (95% CI, 0.15-0.43) suggest that neither a positive or negative result for malignancy is particularly useful for ruling it and ruling it out, though there is uncertainty in the estimates.

Adverse events

No evidence was identified to inform this outcome.

5.3.7. Recommendations

7.

Offer a pancreatic protocol CT scan or magnetic resonance cholangiopancreatography (MRI-MRCP) to people with pancreatic cysts. If more information is needed after one of these tests, offer the other one.

8.

Refer people with any of these high-risk features for resection:

  • obstructive jaundice with cystic lesions in the head of the pancreas
  • enhancing solid component in the cyst
  • a main pancreatic duct that is 10 mm diameter or larger.

9.

Offer EUS after CT and MRI-MRCP if more information on the likelihood of malignancy is needed, or if it is not clear whether surgery is needed.

10.

Consider fine-needle aspiration during EUS if more information on the likelihood of malignancy is needed.

11.

When using fine-needle aspiration, perform carcinoembryonic antigen (CEA) assay in addition to cytology if there is sufficient sample.

12.

For people with cysts that are thought to be malignant, follow the recommendations on staging.

5.3.8. Evidence to recommendations

5.3.8.1. Relative value placed on the outcomes considered

Diagnostic accuracy (sensitivity, specificity, positive predictive value and negative predictive value) and adverse events were considered the critical outcomes for this question. Diagnostic accuracy was reported for all comparisons of interest. Adverse events were only reported for EUS-FNA, MDCT and FDG-PET/CT.

5.3.8.2. Quality of evidence

Evidence was identified on the diagnostic accuracy of CEA, CA 19-9, EUS-FNA, CT, EUS, PET, FDG-PET/CT and MRI. The evidence for CEA ranged from very low to moderate quality, for CA 19-9 was very low, for EUS-FNA ranged from very low to low, for CT was low quality, for EUS ranged from low to moderate quality, for FDG-PET/CT was very low, and for MRI was moderate quality.

The committee noted several limitations with the evidence base. First, a good proportion of the included studies are old and imaging quality is known to have improved since. Second, many of these older studies do not differentiate between IPMN and mucinous cystic neoplasms. Information which is now considered important in identifying which cysts are at higher risk of becoming cancer. Third, there is no validated assay for CEA that is consistently used across all laboratories. This makes it difficult to assess the true diagnostic accuracy of the test. Fourth, the evidence was very fragmented due to different descriptions for malignancy, gold standard of diagnosis, study design and type of cysts.

The committee noted, whilst there was a good amount of data on the diagnostic accuracy of investigations to differentiate mucinous cysts from non-mucinous cysts, there was very little data about what investigations can accurately identify those mucinous cysts which are at high risk of becoming pancreatic cancer. The committee focused on making recommendations about the most effective diagnostic pathway to identify cysts at high risk of becoming malignant as this was the focus of the question.

The committee had more confidence in the quality of evidence from one of the studies related to FDG-PET/CT (Ghaneh et al. 2018) because it was the largest, conducted in a UK NHS setting (and therefore directly applicable) and the study design was judged by the committee to be more robust than that of the other included studies. Therefore in their discussion the committee placed relatively more weight on the evidence from this study than on the rest of the evidence base. Even though the committee believed that the results from this study looked promising (with high specificity and lower yet still relatively good sensitivity), the difficulty is that pancreatic cysts are common and that only those thought to be malignant require further review. The committee also noted that even though the study population was large it only contained a small group of people with pancreatic cysts. Therefore the committee agreed that the evidence from this study was not as applicable for people with pancreatic cysts as for people with jaundice and people without jaundice who have pancreatic abnormalities on imaging.

5.3.8.3. Consideration of clinical benefits and harms

Based on the evidence, the committee noted that MRI had moderate sensitivity and specificity for detecting pancreatic cancer in people with pancreatic cysts. They also noted that whilst CT had low sensitivity, it had high specificity for detecting pancreatic cancer in this population. The committee agreed, based on their knowledge, that both of these investigations are widely available, non-invasive and can provide information on high-risk features of cysts. However they also noted that MRI is more expensive than CT, waiting lists are longer for this investigation and the use of MRI can be contraindicated for some people. Therefore, despite the evidence showing that the sensitivity of CT was not equivalent to that of MRI, the committee recommended either CT or MRI as the initial diagnostic investigation for people with pancreatic cysts in light of the practical constraints around the use of MRI.

Based on their clinical knowledge and experience, the committee noted that if a CT scan is used a pancreatic protocol CT scan should be used to ensure good visualisation of any pathology in the pancreas. They agreed that if MRI is used MRI-MRCP should be used as this will enable the pancreatic duct anatomy to be visualised.

The committee agreed, based on their knowledge, that if the initial CT/MRI identified any high-risk features then the cyst was likely to become malignant so resection would be indicated. They noted that the evidence did not help to identify what the ‘high-risk’ features are. However, they agreed that their recommendation would need to specify them in order to be implementable. The committee agreed the high-risk features that should prompt resection based on their experience and informed by their knowledge of currently accepted definitions.

The committee considered that after an initial CT/MRI there may be some instances where there is uncertainty over whether or not to operate. In these equivocal cases the committee agreed, based on the evidence, that EUS and FNA could help to provide additional information. However, because both EUS and FNA are more invasive, and carry the risk of potential complications, the committee recommended these investigations be reserved for when more information must be obtained in order to determine whether to operate or not.

Although the evidence suggested that FDG-PET/CT may also be helpful in both ruling in and ruling out malignancy of pancreatic cysts, the committee agreed not to recommend its use as it would lead to a very significant increase in costs given the wide variety of cystic lesions and the fact that cysts are relatively commonplace.

The committee also agreed, based on the evidence and their experience, whilst CEA was not helpful in distinguishing between benign and malignant pancreatic cysts, it can provide additional useful diagnostic information. They, therefore, recommended that if an FNA was being done, CEA should be requested at the same time to avoid unnecessary repeat procedures.

The committee agreed that the potential benefits of the recommendations made would be improved and streamlined diagnosis of pancreatic cancer in people with cysts. They considered that EUS/FNA are more invasive investigations and, therefore, are associated with potential complications. They balanced these harms by only recommending the more invasive investigations for a sub-set of people where additional diagnostic information is necessary.

5.3.8.4. Consideration of economic benefits and harms

The committee noted that no relevant published economic evaluations had been identified and no additional economic analysis had been undertaken in this area.

The committee agreed that current practice is to use EUS to investigate most cysts. There should, therefore, be some decrease in costs associated with the recommendations as EUS will now only be used in a sub-set of the population. However, there may also be a corresponding increase in costs associated with the use of the other investigations recommended. The committee agreed that overall the recommendations were likely to be cost neutral.

The committee also noted that although FDG-PET/CT appeared to be very useful in ruling in malignancy and moderately useful in ruling it out. recommending it would have a very significant resource impact. Whilst evidence from the cost utility analysis in Ghaneh et al. (2018; discussed in detail in section 7.5.1) suggested that FDG-PET/CT could be cost effective and cost saving in a patient cohort which included this population the committee acknowledged that malignant cysts only made up 1.5% of the study cohort. The committee also noted, based on their knowledge, that non-malignant pancreatic cysts are otherwise common. The potential number of people that would be eligible for FDG-PET/CT could be large. It would therefore not be appropriate to attach the conclusions of the cost utility study to this subgroup alone. The committee agreed that without stronger evidence of cost effectiveness it could not recommend the use of FDG-PET/CT in the diagnostic pathway of people with pancreatic cysts.

5.3.9. References

  • Ardengh JC, Lopes CV, de Lima LF et al. (2007) Diagnosis of pancreatic tumors by endoscopic ultrasound-guided fine-needle aspiration. World Journal of Gastroenterology 13(22): 3112–6 [PMC free article: PMC4172620] [PubMed: 17589929]
  • Brugge WR, Lewandrowski K, Lee-Lewandrowski E et al. (2004) Diagnosis of pancreatic cystic neoplasms: a report of the cooperative pancreatic cyst study. Gastroenterology 126(5): 1330–6 [PubMed: 15131794]
  • Cao S, Hu Y, Gao X et al. (2016) Serum Carbohydrate Antigen 19-9 in Differential Diagnosis of Benign and Malignant Pancreatic Cystic Neoplasms: A Meta-Analysis. PLoS One 11(11): e0166406 [PMC free article: PMC5105948] [PubMed: 27835676]
  • Cizginer S, Turner BG, Bilge AR et al. (2011) Cyst fluid carcinoembryonic antigen is an accurate diagnostic marker of pancreatic mucinous cysts. Pancreas 40(7): 1024–8 [PubMed: 21775920]
  • Frossard JL, Amouyal P, Amouyal G et al. (2003) Performance of endosonography-guided fine needle aspiration and biopsy in the diagnosis of pancreatic cystic lesions. American Journal of Gastroenterology 98(7): 1516–24 [PubMed: 12873573]
  • Gaddam S, Ge PS, Keach JW et al. (2015) Suboptimal accuracy of carcinoembryonic antigen in differentiation of mucinous and nonmucinous pancreatic cysts: results of a large multicenter study. Gastrointestinal Endoscopy 82(6): 1060–9 [PubMed: 26077458]
  • Gerke H, Jaffe TA, Mitchell RM et al. (2006) Endoscopic ultrasound and computer tomography are inaccurate methods of classifying cystic pancreatic lesions. Digestive and Liver Disease 38(1): 39–44 [PubMed: 16314152]
  • Ghaneh P, Hanson R, Titman A et al. (2018) PET-PANC: multicentre prospective diagnostic accuracy and health economic analysis study of the impact of combined modality 18fluorine-2-fluoro-2-deoxy-d-glucose positron emission tomography with computed tomography scanning in the diagnosis and management of pancreatic cancer. Health Technology Assessment 22(7) [PMC free article: PMC5817411] [PubMed: 29402376]
  • Hirono S, Tani M, Kawai M et al. (2012) The carcinoembryonic antigen level in pancreatic juice and mural nodule size are predictors of malignancy for branch duct type intraductal papillary mucinous neoplasms of the pancreas. Annals of Surgery 255(3): 517–22 [PubMed: 22301608]
  • Jang KM, Kim SH, Min JH et al. (2014) Value of diffusion-weighted MRI for differentiating malignant from benign intraductal papillary mucinous neoplasms of the pancreas. American Journal of Roentgenology 203(5): 992–1000 [PubMed: 25341136]
  • Jin DX, Small AJ, Vollmer CM et al. (2015) A lower cyst fluid CEA cut-off increases diagnostic accuracy in identifying mucinous pancreatic cystic lesions. Journal of Pancreas 16(3): 271–7
  • Kamata K, Kitano M, Omoto S et al. (2016) Contrast-enhanced harmonic endoscopic ultrasonography for differential diagnosis of pancreatic cysts. Endoscopy 48(1): 35–41 [PubMed: 26605974]
  • Kim JH, Eun HW, Park HJ et al. (2012) Diagnostic performance of MRI and EUS in the differentiation of benign from malignant pancreatic cyst and cyst communication with the main duct. European Journal of Radiology 81(11): 2927–35 [PubMed: 22227264]
  • Kim SH, Lee JM, Lee ES et al. (2015) Intraductal papillary mucinous neoplasms of the pancreas: Evaluation of malignant potential and surgical resectability by using MR imaging with MR cholangiography. Radiology 274(3): 723–33 [PubMed: 25302831]
  • Lee HJ, Kim MJ, Choi JY et al. (2011) Relative accuracy of CT and MRI in the differentiation of benign from malignant pancreatic cystic lesions. Clinical Radiology 66: 315–21, 2011 [PubMed: 21356393]
  • Linder JD, Geenen JE, Catalano MF (2006) Cyst fluid analysis obtained by EUS-guided FNA in the evaluation of discrete cystic neoplasms of the pancreas: A prospective single-center experience. Gastrointestinal Endoscopy 64(5): 697–702 [PubMed: 17055859]
  • Moris M, Raimondo M, Woodward TA et al. (2016) Diagnostic Accuracy of Endoscopic Ultrasound-Guided Fine-Needle Aspiration Cytology, Carcinoembryonic Antigen, and Amylase in Intraductal Papillary Mucinous Neoplasm. Pancreas 45(6): 870–5 [PubMed: 26646270]
  • Nagashio Y, Hijioka S, Mizuno N et al. (2014) Combination of cyst fluid CEA and CA 125 is an accurate diagnostic tool for differentiating mucinous cystic neoplasms from intraductal papillary mucinous neoplasms. Pancreatology 14(6): 503–9 [PubMed: 25458668]
  • Nara S, Onaya H, Hiraoka N et al. (2009) Preoperative evaluation of invasive and noninvasive intraductal papillary-mucinous neoplasms of the pancreas: clinical, radiological, and pathological analysis of 123 cases. Pancreas 38(1): 8–16 [PubMed: 18665010]
  • Oh HC, Kang H, Brugge WR (2014) Cyst fluid amylase and CEA levels in the differential diagnosis of pancreatic cysts: a single-center experience with histologically proven cysts. Digestive Diseases and Sciences 59(12): 3111–6 [PubMed: 24965184]
  • Oppong KW, Dawwas MF, Charnley RM et al. (2015) EUS and EUS-FNA diagnosis of suspected pancreatic cystic neoplasms: Is the sum of the parts greater than the CEA? Pancreatology 15(5): 531–7 [PubMed: 26375415]
  • Othman MO, Patel M, Dabizzi E, et al. (2012) Carcino embryonic antigen and long-term follow-up of mucinous pancreatic cysts including intraductal papillary mucinous neoplasm. Digestive and Liver Disease 44: 844–8 [PubMed: 22789399]
  • Pais SA, Attasaranya S, Leblanc JK et al. (2007) Role of endoscopic ultrasound in the diagnosis of intraductal papillary mucinous neoplasms: correlation with surgical histopathology. Clinical Gastroenterology and Hepatology 5(4): 489–95 [PubMed: 17350894]
  • Park WG, Mascarenhas R, Palaez-Luna M et al. (2011) Diagnostic performance of cyst fluid carcinoembryonic antigen and amylase in histologically confirmed pancreatic cysts. Pancreas 40(1): 42–5 [PMC free article: PMC3005131] [PubMed: 20966811]
  • Pitman MB, Genevay M, Yaeger K et al. (2010) High-grade atypical epithelial cells in pancreatic mucinous cysts are a more accurate predictor of malignancy than “positive” cytology. Cancer Cytopathology 118(6): 434–40 [PubMed: 20931638]
  • Pitman MB, Yaeger KA, Brugge WR et al. (2013) Prospective analysis of atypical epithelial cells as a high-risk cytologic feature for malignancy in pancreatic cysts. Cancer Cytopathology 121(1): 29–36 [PubMed: 23132817]
  • Smith AL, Abdul-Karim FW, Goyal A (2016) Cytologic categorization of pancreatic neoplastic mucinous cysts with an assessment of the risk of malignancy: A retrospective study based on the Papanicolaou Society of Cytopathology guidelines. Cancer Cytopathology 124(4): 285–93 [PubMed: 26618476]
  • Song SJ, Lee JM, Kim YJ et al. (2007) Differentiation of intraductal papillary mucinous neoplasms from other pancreatic cystic masses: comparison of multirow-detector CT and MR imaging using ROC analysis. Journal of Magnetic Resonance Imaging 26(1): 86–93 [PubMed: 17659551]
  • Sperti C, Pasquali C, Chierichetti F et al. (2001) Value of 18-fluorodeoxyglucose positron emission tomography in the management of patients with cystic tumors of the pancreas. Annals of Surgery 234(5): 675–80 [PMC free article: PMC1422093] [PubMed: 11685032]
  • Sperti C, Pasquali C, Decet G et al. (2005) F-18-fluorodeoxyglucose positron emission tomography in differentiating malignant from benign pancreatic cysts: a prospective study. Journal of Gastrointestinal Surgery 9(1): 22–8 [PubMed: 15623441]
  • Takanami K, Hiraide T, Tsuda M et al. (2011) Additional value of FDG FDG-PET/CT to contrast-enhanced CT in the differentiation between benign and malignant intraductal papillary mucinous neoplasms of the pancreas with mural nodules. Annals of Nuclear Medicine 25(7): 501–10 [PubMed: 21537945]
  • Talar-Wojnarowska R, Pazurek M, Durko L et al. (2013) Pancreatic cyst fluid analysis for differential diagnosis between benign and malignant lesions. Oncology Letters 5(2): 613–616 [PMC free article: PMC3573134] [PubMed: 23420052]
  • Wu H, Yan LN, Cheng NS et al. (2007) Role of cystic fluid in diagnosis of the pancreatic cystadenoma and cystadenocarcinoma. Hepatogastroenterology 54(79): 1915–8 [PubMed: 18251127]
  • Zhang S, Defrias DV, Alasadi R et al. (2010) Endoscopic ultrasound-guided fine needle aspiration (EUS-FNA): experience of an academic centre in the USA. Cytopathology 21(1): 35–43 [PubMed: 19843142]
  • Zhu H, Jiang F, Zhu J et al. (2017) Assessment of morbidity and mortality associated with EUS-guided FNA for pancreatic cystic lesions: A System Review and Meta-Analysis. Digestive Endoscopy Feb 20

5.3.9.1. Studies included in Cao et al., 2016 (n=13)

  • Fritz S, Hackert T, Hinz U et al. (2011) Role of serum carbohydrate antigen 19–9 and carcinoembryonic antigen in distinguishing between benign and invasive intraductal papillary mucinous neoplasm of the pancreas. British Journal of Surgery 98(1): 104–10 [PubMed: 20949535]
  • Goh BKP, Tan Y, Thng C et al. (2008) How Useful Are Clinical, Biochemical, and Cross-Sectional Imaging Features in Predicting Potentially Malignant or Malignant Cystic Lesions of the Pancreas? Results from a Single Institution Experience with 220 Surgically Treated Patients. Journal of the American College of Surgeons 206(1): 17–27 [PubMed: 18155564]
  • Grobmyer SR, Cance WG, Copeland EM et al. (2009) Is there an indication for initial conservative management of pancreatic cystic lesions? Journal of Surgical Oncology 100(5): 372–74 [PubMed: 19267387]
  • Hirono S, Tani M, Kawai M et al. (2012) The Carcinoembryonic Antigen Level in Pancreatic Juice and Mural Nodule Size Are Predictors of Malignancy for Branch Duct Type Intraductal Papillary Mucinous Neoplasms of the Pancreas. Annals of Surgery 255(3): 517–22 [PubMed: 22301608]
  • Hwang DW, Jang J, Lim C et al. (2011) Determination of Malignant and Invasive Predictors in Branch Duct Type Intraductal Papillary Mucinous Neoplasms of the Pancreas: A Suggested Scoring Formula. Journal of Korean Medical Science 26(6): 740 [PMC free article: PMC3102866] [PubMed: 21655058]
  • Ingkakul T, Sadakari Y, Ienaga J et al. (2010) Predictors of the Presence of Concomitant Invasive Ductal Carcinoma in Intraductal Papillary Mucinous Neoplasm of the Pancreas. Annals of Surgery 2010; 251(1): 70–75 [PubMed: 20009749]
  • Jones NB, Hatzaras I, George N et al. (2009) Clinical factors predictive of malignant and premalignant cystic neoplasms of the pancreas: a single institution experience. HPB 11(8): 664–70 [PMC free article: PMC2799619] [PubMed: 20495634]
  • Kitagawa Y, Unger TA, Taylor S et al. (2003) Mucus is a predictor of better prognosis and survival in patients with intraductal papillary mucinous tumor of the pancreas. Journal of Gastrointestinal Surgery 7(1):12–18 [PubMed: 12559180]
  • Ohtsuka T, Kono H, Nagayoshi Y et al.(2012) An increase in the number of predictive factors augments the likelihood of malignancy in branch duct intraductal papillary mucinous neoplasm of the pancreas. Surgery 151(1): 76–83. [PubMed: 21875733]
  • Sadakari Y, Ienaga J, Kobayashi K et al. (2010) Cyst size indicates malignant transformation in branch duct intraductal papillary mucinous neoplasm of the pancreas without mural nodules. Pancreas 39(2): 232–36 [PubMed: 19752768]
  • Shin SH, Han DJ, Park KT et al. (2010) Validating a Simple Scoring System to Predict Malignancy and Invasiveness of Intraductal Papillary Mucinous Neoplasms of the Pancreas. World Journal of Surgery 34(4): 776–83 [PubMed: 20127242]
  • Sperti C, Bissoli S, Pasquali C et al. (2007) 18-fluorodeoxyglucose positron emission tomography enhances computed tomography diagnosis of malignant intraductal papillary mucinous neoplasms of the pancreas. Annals of Surgery 246(6): 932–37 [PubMed: 18043094]
  • Xu B, Zheng W, Jin D et al. (2011) Predictive Value of Serum Carbohydrate Antigen 19-9 in Malignant Intraductal Papillary Mucinous Neoplasms. World Journal of Surgery 35(5): 1103–09 [PubMed: 21416173]

5.4. People with inherited high risk of pancreatic cancer

Review question: What is the most effective monitoring protocol for adults with an inherited high risk of pancreatic cancer in secondary care to ensure early diagnosis?

5.4.1. Introduction

There are three main groups of people who are at a high risk of developing pancreatic cancer:

  1. those with familial pancreatic cancer
  2. those with hereditary pancreatitis
  3. those with hereditary tumour predisposition syndromes

People with hereditary pancreatitis have a 70 fold increased risk of pancreatic cancer. The life time risk is 35-40% and rises with age. People with familial pancreatic cancer have a life time risk of 30-50% which rises with age.

Guidance is needed on the most effective monitoring protocol to ensure early diagnosis in people with an inherited high risk of pancreatic cancer.

5.4.1.1. Review protocol summary

The review protocol summary used for this question can be found in Table 58. Full details of the review protocol can be found in Appendix C.

Table 53. Clinical review protocol summary for the review of most effective monitoring protocol for adults with an inherited high risk of pancreatic cancer.

Table 53

Clinical review protocol summary for the review of most effective monitoring protocol for adults with an inherited high risk of pancreatic cancer.

5.4.2. Description of clinical evidence

Eighteen articles were identified: 17 of these concerned screening/surveillance programs, whilst 1 was a secondary study that reported on the psychological burden/quality of life of participating in 1 of these screening programs. All 17 of the primary studies reported diagnostic yield (early diagnosis). A summary of the included studies is presented in Table 54.

Seventeen studies (n=2661) were identified that evaluated the diagnostic performance of screening and/or surveillance programs for adults with an inherited ‘high’ risk of pancreatic cancer: 5 prospective cohort studies (Canto et al. 2006; Chang et al. 2017; Potjer et al. 2013; Vasen et al. 2016; Verna et al. 2010), 1 retrospective review of a prospective cohort study (Nocholson et al. 2015), and 11 case series (Al-Sukhni et al. 2012; Bartsch et al. 2016; Canto et al. 2004; Canto et al. 2012; Del Chiaro et al. 2015; Harinck et al. 2016; Kimmey et al. 2002; Ludwig et al. 2011; Poley et al. 2009; Sud et al. 2014; Zubarik et al. 2011). The majority of the studies included familial pancreatic cancer (FPC), which was typically defined as an individual that has two or more relatives with pancreatic cancer. In addition, all of the studies (with the exception of Canto et al. 2012 and Harinck et al. 2016) consisted of an initial test(s) and, given an abnormal result, subsequent imaging or other tests. The most common initial test (11 studies) was MRI/MRCP, or MRI combined with EUS±FNA, whilst the most common subsequent test was EUS±FNA. Only two studies (Canto et al. 2006; Canto et al. 2012) used CT as part of the initial screening test and in both cases this was in combination with other tests (EUS and/or MRI). One multicentre prospective study (n=546; Zubarik et al. 2011) used serum CA 19-9 as the initial test and EUS-FNA given an abnormal result (values >37 U/ml). Data on the diagnostic yield and adverse events of screening/surveillance programs is not amenable to a meta-analysis or depiction using forest plots (however see Nicholson et al. 2015 below). Therefore a narrative summary and table listing the relevant results have been presented.

One retrospective review of a prospective cohort study (n=60; Nicholson et al. 2015) examined the incidence of post-ERCP pancreatitis with and without prophylaxis in people with familial pancreatic cancer or hereditary pancreatitis.

One interrupted time series study (n=152; Konings et al. 2016) examined participants enrolled in the annual surveillance program reported in Harinck et al. 2016 (see above). Although this secondary study did not report health-related quality of life, it reported change on the Cancer Worry scale and the HADS-Anxiety and HADS–Depression scales and so was included.

The QUADAS-2 checklist was used to evaluate the risk of bias and applicability (indirectness) of the screening/surveillance studies. Due to the type of data (diagnostic yield) reported, the criteria of inconsistency and imprecision were not evaluated for these studies, and the quality of each study was therefore rated individually. A narrative summary of the evidence is presented. The GRADE risk of bias tool was used to evaluate 1 study that reported post-ERCP pancreatitis with and without prophylaxis.

Further information about the search strategy can be found in Appendix D. See study selection flow chart in Appendix E, forest plots in Appendix H, summary of QUADAS-2 study quality evaluations in Appendix J, study evidence tables in Appendix F and list of excluded studies in Appendix G.

5.4.3. Summary of included studies

A summary of the studies that were included in this review is presented in Table 54.

Table 54. Summary of included studies.

Table 54

Summary of included studies.

5.4.4. Clinical evidence profile

5.4.4.1. Screening/surveillance studies

5.4.4.1.1. Narrative summary of evidence

The majority of the 17 studies were in adults with familial pancreatic cancer, the majority of which also included relatively small numbers of individuals with identified germline mutations such as BRCA, p16 or p53. The majority of the participants were female, ranging from 55% to 75% of the samples (approximately 60% female across 15 studies). One study did not report patient characteristics, and in 1 study this information was unclear. Nine studies were conducted in the USA/Canada, 6 in Europe (2 of which were international multicentre studies), and 1 in Taiwan. Only 1 study was conducted in the UK (Nicholson et al. 2015).

The most common initial screening test in the 17 published studies was MRI/MRCP with or without additional EUS (8 studies), whilst the most common test given an abnormal initial result was EUS±FNA (10 studies). Three screening programs did not use a subsequent test given an abnormal result. Fifteen of the articles included only individuals with at least a 5% or more increased risk of pancreatic cancer compared to those in the normal population, whilst two of the studies included individuals at ‘average’ risk of pancreatic cancer.

The diagnostic yield reported in the identified screening/surveillance studies varied widely, ranging from 0.9% to 39%, depending on the type of malignant or premalignant lesion identified, the population and reference test (e.g. surgical pathology only) employed, whether additional tests were conducted given initial abnormal results, and whether results included baseline results only or included follow up.

Of the 2661 individuals at risk, 2418 were screened: 41 (1.7%) of these were diagnosed with pancreatic cancer, resulting in an overall screening efficiency of 59 screened individuals to detect 1 case of pancreatic cancer. If individuals with premalignant lesions are included (i.e. those with IPMN and/or PanIN≥2), 145 individuals (including those with pancreatic cancer) were identified, resulting in a screening efficiency of 6.0% (1 malignant or premalignant lesions for every 17 individuals at risk screened). This suggests that screening high- and moderate- individuals at risk for malignant lesions only will be both costly and time consuming and that screening programs should include premalignant lesions.

Only 1 study (Vasen et al. 2016), which evaluated the diagnostic yield of MRI/MRCP, reported overall survival (a 5-year overall survival of 24% for the CDKN2A/p16 cohort with pancreatic ductal adenocarcinoma). Very few adverse events as a result of participating in the screening/surveillance programs were reported in the 13 studies that reported procedure-related complications. The majority of these were reported in 1 study (Canto et al. 2006) or were related to post-ERCP pancreatitis. Although no studies were found that reported health-related quality of life, there was 1 secondary study (Konings et al. 2016) related to participation in the screening/surveillance program reported in Harinck et al. 2016 (comprising EUS and MRI), that reported significant decreases in worry associated with having cancer (approximately 0.5 point decrease on the Cancer Worry Scale) for every year enrolled in the program. However, participants in this study reported no significant change in depression and anxiety.

The risk of bias and indirectness for each study was generally low for both quality measures with the exception of 2 studies (Canto et al. 2012; Ludwig et al. 2011) both of which had an unclear risk of bias. Overall, the majority of the studies were of ‘high’ quality (rated as ++), with the aforementioned 2 studies rated as ‘low’ (+) quality. Generally it was not clear whether the reference test(s) was interpreted without knowledge of the index test(s) results.

A summary of the evidence for this review question is presented in Table 55.

Table 55. Summary of evidence and quality evaluation.

Table 55

Summary of evidence and quality evaluation.

5.4.4.2. ERCP with prophylaxis versus ERCP only

Table 56. Summary clinical evidence profile for ERCP with prophylaxis versus ERCP only on reducing post-ERCP pancreatitis in people at high risk of pancreatic cancer.

Table 56

Summary clinical evidence profile for ERCP with prophylaxis versus ERCP only on reducing post-ERCP pancreatitis in people at high risk of pancreatic cancer.

5.4.5. Economic evidence

A literature review of published cost effectiveness analyses did not identify any relevant studies for this topic. Although there were potential implications for resource use associated with making recommendations in this area, other topics in the guideline were agreed as a higher economic priority. Consequently, bespoke economic modelling was not done for this topic.

5.4.6. Evidence Statements

5.4.6.1. Screening/surveillance studies

Diagnostic yield

There was inconsistent evidence from 17 prospective cohort studies (n=2661) on the diagnostic yield – i.e. early diagnosis or identification of malignant and premalignant pancreatic lesions - of pancreatic cancer screening/surveillance programs in high- and moderate- risk adults. Although the majority of the studies reporting the results of these programs were of high (++) quality and used pathological diagnosis, the diagnostic yield was highly variable, ranging from 0.9% to 39%. This variability is likely dependent on the initial index tests on the subgroups (e.g. breast cancer susceptibility gene, p16, p53) and types of lesion included in the samples recruited by the programs. The overall screening efficiency of the programs, which were mainly conducted in the USA, in detecting pancreatic cancer was 1.7% (1 detected case of pancreatic cancer for every 59 individuals at risk screened or monitored) and 6.0% if premalignant lesions (IPMN and PanIN≥2) are included (1 detected case for every 16 individuals at risk screened or monitored).

Overall survival

No evidence was identified to inform this outcome.

Adverse events

Eleven high (++) quality and 2 low (+) quality prospective cohort studies (n=1329) indicated that the incidence of adverse events related to the tests used in the screening/surveillance programs of high- and moderate-risk individuals was very low (<1% excluding post-ERCP pancreatitis). The majority of the reported adverse events – 22 cases of post-test abdominal pain (of 78 participants), and 5 cases of post-ERCP pancreatitis (of 65 participants) - were from 1 ‘high’ (++) quality study (Canto 2006) that combined EUS with CT as either the initial index test or subsequent test given an initial abnormal finding. In the 3 studies (excluding Nicholson 2015; see below) that utilised ERCP, there were 7 cases of post-ERCP pancreatitis (5.9%) out of the 119 participants that received it.

5.4.6.2. ERCP with prophylaxis vs ERCP only

Adverse events

Very low quality evidence from 1 single centre prospective cohort study (n=48, 56 ERCP procedures) showed that there is a clinically important difference favouring ERCP with prophylaxis on reducing post-ERCP pancreatitis in people with familial pancreatic cancer compared to ERCP without prophylaxis: RR 0.34 (95%CI, 0.14-0.86).

Very low quality evidence from 1 single centre prospective cohort study (n=12, 24 ERCP procedures) showed no clinically important difference between ERCP with prophylaxis and ERCP without prophylaxis in people with hereditary pancreatitis (there were no cases in either group).

5.4.7. Recommendations

13.

Ask people with pancreatic cancer if any of their first-degree relatives has had it. Address any concerns the person has about inherited risk.

14.

Offer surveillance for pancreatic cancer to people with:

  • hereditary pancreatitis and a PRSS1 mutation
  • BRCA1, BRCA2, PALB2, or CDKN2A (p16) mutations, and one or more first-degree relatives with pancreatic cancer
  • Peutz–Jeghers syndrome.

15.

Consider surveillance for pancreatic cancer for people with:

  • 2 or more first-degree relatives with pancreatic cancer, across 2 or more generations
  • Lynch syndrome (mismatch repair gene [MLH1, MSH2, MSH6, or PMS2] mutations) and any first-degree relatives with pancreatic cancer.

16.

Consider an MRI-MRCP or EUS for pancreatic cancer surveillance in people without hereditary pancreatitis.

17.

Consider a pancreatic protocol CT scan for pancreatic cancer surveillance in people with hereditary pancreatitis and a PRSS1 mutation.

18.

Do not offer EUS to detect pancreatic cancer in people with hereditary pancreatitis.

5.4.8. Evidence to recommendations

5.4.8.1. Relative value placed on the outcomes considered

Early diagnosis, survival, diagnostic accuracy (including sensitivity, specificity, positive predictive value and negative predictive value), adverse events of interventions and health related quality of life were considered to be the critical outcomes for this question.

Diagnostic yield was reported for all studies and adverse events were reported for the majority of studies. Overall survival was only reported by one study and early diagnosis and health-related quality of life were not reported.

5.4.8.2. Quality of evidence

The QUADAS-2 checklist was used to evaluate the risk of bias and applicability of the screening or surveillance studies. Due to the type of data reported (diagnostic yield), the criteria of inconsistency and imprecision were not evaluated for the screening or surveillance studies. The GRADE risk of bias tool was used to evaluate the study that reported post- ERCP pancreatitis with and without prophylaxis.

For screening or surveillance, there were high quality studies for diagnostic yield and overall survival. The studies reporting adverse events were mostly high quality but with two low quality studies. For ERCP with prophylaxis versus ERCP only, there was only low quality evidence on adverse events.

5.4.8.3. Consideration of clinical benefits and harms

Based on their clinical knowledge, the committee noted that 5-10% of cases of pancreatic cancer are caused by hereditary factors. Consequently they agreed that it was very important to discuss family history with everyone who has pancreatic cancer so that people who have any hereditary factors can be identified earlier.

The committee noted, based on the evidence, that there are certain groups of hereditary factors that carry a higher risk of developing pancreatic cancer (an affected individual with hereditary pancreatitis with a PRSS1 mutation; people who are BRCA1, BRCA2, PALB2 or CDKN2A (p16) mutation carriers with one or more affected first-degree relatives with pancreatic cancer; people with Peutz–Jeghers syndrome, regardless of family history). The committee acknowledged that the data on survival were too limited to prove there is a survival benefit of surveillance in these people. However, they noted the data from Vasen et al (2016), who had surveilled individuals at high risk of pancreatic cancer, reported an overall resection rate of 75% and overall survival at 5 years of 24% compared to a resection rate of 15% and 5-year survival rate of 4-7% for patients with sporadic symptomatic pancreatic ductal adenocarcinoma. Since these figures are higher than what would normally be expected for people with pancreatic cancer, the committee agreed these data were suggestive that surveillance could confer benefits to survival outcomes.

The committee also noted that these hereditary factors are usually associated with very poor prognosis which can cause a lot of anxiety to the people who have them. The committee considered that offering surveillance to those people with hereditary factors that carry a higher risk of developing pancreatic cancer, would help to resolve this anxiety. They also agreed, based on their experience, that surveillance of these people should lead to earlier diagnosis of pancreatic cancer and earlier treatment, which will help to improve the experience of patients. They therefore agreed to recommend that people with these hereditary factors should be offered surveillance for pancreatic cancer.

The committee also noted there are other groups of hereditary factors that carry an increased risk of developing pancreatic cancer, but which are not classified as ‘high risk’. The committee agreed that there were likely to be benefits of surveillance for these people for pancreatic cancer but the balance was less clear. They therefore agreed a weaker recommendation for surveillance in people with first-degree relatives (FDRs) with pancreatic cancer from a familial pancreatic cancer kindred with at least 2 FDRs in 2 or more generations; people with mismatch repair gene (MLH1, MSH2, MSH6, PMS2) mutations (Lynch syndrome) and one affected FDR with pancreatic cancer. This would be consistent with the current EUROPAC registry entry requirements (unpublished) and the International Cancer of the Pancreas Screening (CAPS) Consortium consensus statement on inherited risk (Canto et al. 2013).

The committee agreed that the evidence on the diagnostic yield of CT, MRI and EUS in surveillance had shown they were all accurate at identifying early tumours. However, from the available evidence the committee could not identify which of these investigations was the most effective. Given this uncertainty, the committee recommended further research to evaluate the surveillance tests and frequency of surveillance that produce the greatest diagnostic yield and overall surveillance efficiency. The Committee also noted that repeated CT scanning would expose people to harms associated with radiation and therefore did not want to recommend this as an option for people without hereditary pancreatitis in whom a larger percentage of people would have a relatively smaller risk. However, they agreed that a pancreatic protocol CT scan, for pancreatic cancer surveillance should be considered for people with hereditary pancreatitis and a PRSS1 mutation who would be at higher risk of developing pancreatic cancer.

Based on their clinical knowledge and experience, the committee noted that if a CT scan is used (in people with hereditary pancreatitis) a pancreatic protocol CT scan would be needed to ensure good visualisation of any pathology in the pancreas. They also agreed that if MRI is used MRI-MRCP should be used as this will enable the pancreatic duct anatomy to be visualised.

The committee noted, based on their knowledge and experience, that the fibrosis, distortion and calcium deposits caused by hereditary pancreatitis prevent the detection of small pancreatic tumours by EUS. They therefore agreed that EUS should not be used to detect pancreatic cancer if the person has hereditary pancreatitis.

The committee noted that the data had shown ERCP with prophylaxis was better at reducing post-ERCP pancreatitis in people with familial pancreatic cancer, compared to ERCP without prophylaxis. However, given that the evidence was from a single, very low quality study the committee agreed not to make a recommendation about this intervention.

The committee agreed that the potential benefits of the recommendations made would be more directed and integrated management of people with hereditary factors, improved detection of pre-malignant lesions and potential improvements in survival. They noted that the recommendations for surveillance had the potential to both increase and decrease anxiety of the person; knowing you are at high risk of developing pancreatic cancer may increase anxiety which would hopefully be offset by being offered surveillance. However, anxiety may also increase around the time that the surveillance occurs as you wait to find out if you have developed pancreatic cancer or not. On balance, the committee agreed that the potential benefits outweighed the harms.

5.4.8.4. Consideration of economic benefits and harms

The committee noted that no relevant published economic evaluations had been identified and no additional economic analysis had been undertaken in this area.

Extending surveillance to individuals with two blood relatives affected by pancreatic cancer would lead to an increase in resource use through increased imaging and health care practitioners time. However, as for other recommendations in this area, only a small proportion of people have an inherited elevated risk of developing pancreatic cancer and consequently the overall resource impact would be small. It was also noted that surveillance of these high risk individuals could lead to earlier intervention improving quality of life and avoiding costs of adverse events and complications.

The committee agreed that the recommendations made were unlikely to have a significant resource impact due to the small number of people who have an inherited risk of developing pancreatic cancer.

5.4.9. Research recommendations

1.

Research should be undertaken to evaluate the most clinically effective and cost effective initial surveillance tests, additional tests and frequency of surveillance that produce the greatest diagnostic yield and overall surveillance efficiency.

At the present time we do not know what the best initial surveillance and subsequent tests are, nor the frequency of the surveillance that will produce the best diagnostic yield for people with an inherited high risk of pancreatic cancer, whilst maintaining quality of life. These will depend upon the accuracy of the tests available, the level of risk and the rate at which the risk materialises.

Individuals with an inherited risk of pancreatic cancer have a highly variable risk dependent on their particular genotype, each with a widely differing levels of risk, or the particular phenotype each also with a variable level of risk. In each case there is a threshold of risk and frequency of testing that would need to be determined to make surveillance effective.

5.4.10. References

  • Al-Sukhni W, Borgida A, Rothenmund H et al. (2012) Screening for pancreatic cancer in a high-risk cohort: an eight-year experience. Journal of Gastrointestinal Surgery 16(4): 771–783 [PubMed: 22127781]
  • Bartsch DK, Slater EP, Carrato A et al. (2016) Refinement of screening for familial pancreatic cancer. Gut 65(8): 1314–1321 [PubMed: 27222532]
  • Canto MI, Goggins M, Hruban RH et al. (2006) Screening for early pancreatic neoplasia in high-risk individuals: a prospective controlled study. Clinical Gastroenterology and Hepatology 4(6): 766–781 [PubMed: 16682259]
  • Canto MI, Goggins M, Yeo CJ et al. (2004). Screening for pancreatic neoplasia in high-risk individuals: an EUS-based approach. Clinical Gastroenterology and Hepatology 2(7): 606–621 [PubMed: 15224285]
  • Canto, MI, Harink, F, Hruban, RH et al. (2013). International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut 62(3): 339–347 [PMC free article: PMC3585492] [PubMed: 23135763]
  • Canto MI, Hruban RH, Fishman EK et al. (2012) Frequent detection of pancreatic lesions in asymptomatic high-risk individuals. Gastroenterology 142(4): 796–804. [PMC free article: PMC3321068] [PubMed: 22245846]
  • Chang MC, Wu CH, Yang SH et al. (2017) Pancreatic cancer screening in different risk individuals with family history of pancreatic cancer-a prospective cohort study in Taiwan. American Journal of Cancer Research 7(2): 357 [PMC free article: PMC5336508] [PubMed: 28337383]
  • Del Chiaro M, Verbeke CS, Kartalis N et al. (2015) Short-term Results of a Magnetic Resonance Imaging–Based Swedish Screening Program for Individuals at Risk for Pancreatic Cancer. JAMA Surgery 150(6): 512–518 [PubMed: 25853369]
  • Harinck F, Konings IC, Kluijt I et al. (2016) A multicentre comparative prospective blinded analysis of EUS and MRI for screening of pancreatic cancer in high-risk individuals. Gut 65(9): 1505–1513 [PubMed: 25986944]
  • Kimmey MB, Bronner MP, Byrd DR et al. (2002) Screening and surveillance for hereditary pancreatic cancer. Gastrointestinal Endoscopy 56(4): S82–S86 [PubMed: 12297755]
  • Konings IC, Sidharta GN, Harinck, F et al. (2015) Repeated participation in pancreatic cancer surveillance by high – risk individuals imposes low psychological burden. Psycho – Oncology 25(8): 971–978 [PubMed: 26632416]
  • Ludwig E, Olson SH, Bayuga S et al. (2011) Feasibility and yield of screening in relatives from familial pancreatic cancer families. The American Journal of Gastroenterology 106(5): 946–954 [PMC free article: PMC3683863] [PubMed: 21468009]
  • Nicholson JA, Greenhalf W, Jackson R et al. (2015) Incidence of post-ERCP pancreatitis from direct pancreatic juice collection in hereditary pancreatitis and familial pancreatic cancer before and after the introduction of prophylactic pancreatic stents and rectal diclofenac. Pancreas 44(2): 260–265 [PubMed: 25438071]
  • Poley JW, Kluijt I, Gouma DJ et al. (2009) The yield of first-time endoscopic ultrasonography in screening individuals at a high risk of developing pancreatic cancer. The American Journal of Gastroenterology 104(9): 2175–2181 [PubMed: 19491823]
  • Potjer TP, Schot I, Langer P et al. (2013) Variation in precursor lesions of pancreatic cancer among high-risk groups. Clinical Cancer Research 19(2): 442–449. [PubMed: 23172884]
  • Sud A, Wham D, Catalano M et al. (2014) Promising outcomes of screening for pancreatic cancer by genetic testing and endoscopic ultrasound. Pancreas 43(3): 458–461 [PubMed: 24622079]
  • Vasen H, Ibrahim I, Ponce CG et al. (2016) Benefit of surveillance for pancreatic cancer in high-risk individuals: outcome of long-term prospective follow-up studies from three European expert centers. Journal of Clinical Oncology 34(17): 2010–2019 [PubMed: 27114589]
  • Verna EC, Hwang C, Stevens PD et al. (2010) Pancreatic cancer screening in a prospective cohort of high-risk patients: a comprehensive strategy of imaging and genetics. Clinical Cancer Research 16(20): 5028–5037 [PubMed: 20876795]
  • Zubarik R, Gordon SR, Lidofsky, SD et al. (2011) Screening for pancreatic cancer in a high-risk population with serum CA 19-9 and targeted EUS: a feasibility study. Gastrointestinal Endoscopy 74(1): 87–95 [PubMed: 21704809]
Copyright © NICE 2018.
Bookshelf ID: NBK536656

Views

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...