A Large-Scale Proteomics Resource of Circulating Extracellular Vesicles for Biomarker Discovery in Pancreatic Cancer

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    The authors analyze a comprehensive cohort of human plasma samples to identify an extracellular vesicles protein signature for earlier diagnosis of pancreatic cancer. Application of liquid biopsies is valuable, and it addresses a significant clinical problem as pancreas cancer is often diagnosed in late stage with standard biopsies often nondiagnostic. The strength of evidence is largely solid; however, certain aspects of the data were deemed incomplete, given the broad manuscript claims. This work supports the use of extracellular vesicles in clinical settings, with potential interest to scientists and clinicians.

This article has been Reviewed by the following groups

Read the full article

Abstract

Pancreatic cancer has the worst prognosis of all common tumors. Earlier cancer diagnosis could increase survival rates and better assessment of metastatic disease could improve patient care. As such, there is an urgent need to develop biomarkers to diagnose this deadly malignancy. Analyzing circulating extracellular vesicles (cEVs) using ‘liquid biopsies’ offers an attractive approach to diagnose and monitor disease status. However, it is important to differentiate EV-associated proteins enriched in patients with pancreatic ductal adenocarcinoma (PDAC) from those with benign pancreatic diseases such as chronic pancreatitis and intraductal papillary mucinous neoplasm (IPMN). To meet this need, we combined the novel EVtrap method for highly efficient isolation of EVs from plasma and conducted proteomics analysis of samples from 124 individuals, including patients with PDAC, benign pancreatic diseases and controls. On average, 912 EV proteins were identified per 100µL of plasma. EVs containing high levels of PDCD6IP, SERPINA12 and RUVBL2 were associated with PDAC compared to the benign diseases in both discovery and validation cohorts. EVs with PSMB4, RUVBL2 and ANKAR were associated with metastasis, and those with CRP, RALB and CD55 correlated with poor clinical prognosis. Finally, we validated a 7-EV protein PDAC signature against a background of benign pancreatic diseases that yielded an 89% prediction accuracy for the diagnosis of PDAC. To our knowledge, our study represents the largest proteomics profiling of circulating EVs ever conducted in pancreatic cancer and provides a valuable open-source atlas to the scientific community with a comprehensive catalogue of novel cEVs that may assist in the development of biomarkers and improve the outcomes of patients with PDAC.

Article activity feed

  1. eLife assessment

    The authors analyze a comprehensive cohort of human plasma samples to identify an extracellular vesicles protein signature for earlier diagnosis of pancreatic cancer. Application of liquid biopsies is valuable, and it addresses a significant clinical problem as pancreas cancer is often diagnosed in late stage with standard biopsies often nondiagnostic. The strength of evidence is largely solid; however, certain aspects of the data were deemed incomplete, given the broad manuscript claims. This work supports the use of extracellular vesicles in clinical settings, with potential interest to scientists and clinicians.

  2. Reviewer #1 (Public Review):

    In this manuscript, Bockorny, Muthuswamy, and Huang et al. performed proteomics analysis of plasma extracellular vesicles (EVs) from pancreatic ductal adenocarcinoma (PDAC) patients and patients with benign pancreatic diseases (chronic pancreatitis and intraductal papillary mucinous neoplasm, IPMN) to develop a 7-EV protein signature that predicts PDAC. Moveover, the authors identified PSMB4, RUVBL2, and ANKAR as being associated with metastasis. These studies provide important insight into alterations of EVs during PDAC progression and the data supporting predict PDAC with EV protein signatures are solid. However, there are certain concerns regarding the rigor and novelty of the data analysis and interpretation, as well as the clinical implications, as detailed below.

    1. Plasma EVs were characterized by transmission electron microscopy and nanoparticle tracking analysis to confirm their morphology and size. The authors should also include an analysis of putative EV markers (e.g., tetraspanins, syntenin, ALIX, etc.) to confirm that the analyzed particles are EVs.

    2. The authors identified multiple over-expressed proteins in PDAC based on their fold change and p-value; however, due to the heterogeneity of PDAC, it is necessary to show a heatmap displaying their abundance in all samples. High fold change does not necessarily indicate consistently high abundance in all PDAC samples.

    3. PSMB4, RUVBL2, and ANKAR were identified as being associated with metastasis. The authors state that they intended to distinguish early and late-stage cancer samples, but it is unclear why they chose to compare metastatic and non-metastatic samples, as the non-metastatic group also includes late-stage cancer samples. This sentence should be rephrased to more accurately reflect the sample types profiled.

    4. Non-metastatic and metastatic patients were separated based on global protein abundance. The samples within each group display significant heterogeneity with, some samples displaying similar patterns although they were classified into different groups (Figure 3A), and the samples within the same group, particularly the metastasis group, did not consistently exhibit similar patterns of protein abundance. The authors should clarify this point.

    5. The authors performed the survival analysis on a set of EV proteins but did not specify the origin of these markers or how many markers were examined. The authors should show their abundances across different groups, such as different stages and metastasis status.

    6. The classification model yielded a 100% accuracy, which may refer to AUC, in their discovery cohort, but it decreased to 89% in the independent cohort. This suggests that the authors have encountered overfitting issues with their model, where it performed well on the discovery cohort but did not generalize well to the independent cohort. The authors should clarify this point. The AUC score of the 7-EV signature is 0.89 and is not equivalent to prediction accuracy. In order to demonstrate prediction accuracy, the authors should show the confusion matrix of training and testing data as well as other evaluation metrics, such as accuracy, precision, and recall.

    7. The authors should include more details of their model and the process of selection of signatures to enhance the reproducibility and transparency of their methods.

  3. Reviewer #2 (Public Review):

    The authors intended to identify a protein signature in extracellular vesicles of serum to distinguish pancreatic ductal adenocarcinoma from benign pancreatic diseases.

    A major strength of the work presented is the valuable profiling of a significant number of patient samples, with a rich cohort of patients with pancreatic cancer, benign pancreatic diseases, and healthy controls. However, despite the strong cohorts presented, the numbers of patient samples for benign pancreatic diseases as well as controls were very limited.

    Also, the method used to isolate vesicles, EVTrap, recognizes double bilayers, which means that it can detect cellular debris and apoptotic bodies, which are very common in the circulation of patients that are undergoing chemotherapy. It would be important to identify the patients that are therapy naïve and the ones that are not because of this possible bias. Additionally, the transmission electron microscopy data reflect this heterogeneity of the samples, also with little identification of double bilayered vesicles. It would be important to identify some extracellular vesicles markers in those preparations to strengthen the quality of the samples analysed. What is more, previously published work with this same methodology identifies around 2000 proteins per sample. It would be important to explain why in this study there seems to be a reduction in more than 50% of the amount of proteins identified in the vesicles.

    One of the proteins that constantly surges on the analysis is KRT20. It would be important to proceed with the analysis by first filtering out possible contaminants of the proteomics, of which keratins are the most common ones. Finally, none of the 7-extracellular vesicle protein signatures has been validated by other techniques, such as western blot, in extracellular vesicles isolated by other, standard, methods, such as size exclusion chromatography.

    A distinct technique for protein analysis was done but not a different method of isolation of these vesicles. This would strengthen the results and the origin of the proteins.

    The conclusions that are reached do not fully meet the proposed aims of the identification of a protein signature in circulating extracellular vesicles that could improve early detection of the disease. The authors did not demonstrate the superiority of detection of these proteins in extracellular vesicles versus simply performing an ELISA, nor their superiority with respect to the current standard procedure for diagnosis.

    The authors also suggest that profiling of circulating extracellular vesicles provides unique insights into systemic immune changes during pancreatic cancer development. How is this better than a regular hemogram is not clear.

    Finally, it would be important to determine how this signature compares with many others described in the literature that have the exact same aim. Why and how would this one be better?

  4. Reviewer #3 (Public Review):

    This work investigates the use of extracellular vesicles (EVs) in blood as a noninvasive 'liquid biopsy' to aid in the differentiation of patients with pancreatic cancer (PDAC) from those with benign pancreatic disease and healthy controls, an important clinical question where biopsies are frequently non-diagnostic. The use of extracellular vesicles as biomarkers of disease has been gaining interest in recent history, with a variety of published methods and techniques, looking at a variety of different compositions ('the molecular cargo') of EVs particularly in cancer diagnosis (Shah R, et al, N Engl J Med 2018; 379:958-966).

    This study adds to the growing body of evidence in using EVs for earlier detection of pancreatic cancer, identifying both new and known proteins of interest. Limitations in studying EVs, in general, include dealing with low concentrations in circulation and identifying the most relevant molecular cargo. This study provides validation of assaying EVs using the novel EVtrap method (Extracellular Vesicles Total Recovery And Purification), which the authors show to be more efficient than current standard techniques and potentially more scalable for larger clinical studies.

    The strength of this study is in its numbers - the authors worked with a cohort of 124 cases, 93 of them which were PDAC samples, which are considered large for an EV study (Jia, E et al. BMC Cancer 22, 573 (2022)). The benign disease group (n=20, between chronic pancreatitis and IPMNs) and healthy control groups (n=11) were relatively small, but the authors were not only able to identify candidate biomarkers for diagnosis that clearly stood out in the PDAC cohort, but also validate it in an independent cohort of 36 new subjects.

    Proteins they have identified as associated with pancreatic cancer over benign disease included PDCD6IP, SERPINA12, and RUVBL2. They were even able to identify a set of EV proteins associated with metastasis and poorer prognosis, which include the proteins PSMB4, RUVBL2 and ANKAR and CRP, RALB and CD55. Their 7-EV protein signature yielded an 89% prediction accuracy for the diagnosis of PDAC against a background of benign pancreatic diseases that is compelling and comparable to other studies in the literature (Jia, E. et al. BMC Cancer 22, 573 (2022)).

    The limitations of this study are its containment within a single institution - further studies are warranted to apply the authors' 7-EV protein PRAC panel to multiple other cases at other institutions in a larger cohort.