Registered report: Melanoma genome sequencing reveals frequent PREX2 mutations

  1. Denise Chroscinski
  2. Darryl Sampey
  3. Alex Hewitt
  4. Reproducibility Project: Cancer Biology  Is a corresponding author
  1. Noble Life Sciences, United States
  2. BioFactura, United States
  3. University of Melbourne, Australia

Decision letter

  1. Roger Davis
    Reviewing Editor; University of Massachusetts Medical School, United States

eLife posts the editorial decision letter and author response on a selection of the published articles (subject to the approval of the authors). An edited version of the letter sent to the authors after peer review is shown, indicating the substantive concerns or comments; minor concerns are not usually shown. Reviewers have the opportunity to discuss the decision before the letter is sent (see review process). Similarly, the author response typically shows only responses to the major concerns raised by the reviewers.

Thank you for sending your work entitled “Registered report: Melanoma genome sequencing reveals frequent PREX2 mutations” for consideration at eLife. Your article has been favorably evaluated by Charles Sawyers (Senior editor) and 4 reviewers, one of whom is a member of our Board of Reviewing Editors.

The Reviewing editor and the other reviewers discussed their comments before we reached this decision, and the Reviewing editor has assembled the following comments to help you prepare a revised submission.

There are two main conclusions drawn in the Nature paper by Berger et al. First, PREX2 is identified as a frequently (14%) non-synonymously mutated gene in melanoma. Second, that ectopic expression of cancer-associated PREX2 proteins promote melanoma genesis in a xenograft assay in mice. This is an important study because the identification of driver (vs passenger) mutations in cancer is critical for understanding the mechanism of tumorigenesis.

The first conclusion is addressed by Chroscinski and colleagues in the literature summary. The authors note that validation of this conclusion is not supported by several studies, including meta-analysis. However, Chroscinski and colleagues state that two papers support the conclusion that PREX2 is frequently mutated in melanoma. This is not correct. Turajlic et al. do describe a PREX2 mutation, but the analysis is limited to a single patient, while Furney et al. only refer to PREX2 mutations described by Berger et al. Thus, these papers do not support the conclusion that PREX2 is frequently mutated in melanoma. It also appears to be significant that another study of melanoma exome sequencing by many of the same authors published 2 months after Berger et al. (Hodis et al.) does not appear to identify PREX2 as a frequently mutated gene. Together, these considerations do not provide support for the conclusion presented by Berger et al. that PREX2 is frequently mutated in melanoma. This needs to be more directly addressed by Chroscinski and colleagues: they should discuss statistical thresholds for calling mutations significant (e.g., MutSig) and examine PREX2 mutations in public cancer genome portals such as cBioPortal (MSK) or the Broad Tumor Portal algorithms (in addition to reporting on the negative studies).

The second conclusion (that is addressed by re-analysis by Chroscinski and colleagues) is important: that PREX2 is a driver mutation rather than a passenger mutation during melanoma formation. The proposed experimental design appears to replicate the original study. However, a number of issues were raised by the reviewers:

1) The PREX2 mutations reported by Berger et al. are present throughout the PREX2 sequence. This is more consistent with passenger mutation than driver mutation. The restriction of the re-analysis to a limited number of mutations originally examined is therefore problematic. To document that the PREX2 mutations acts as drivers, it would be best to examine the same mutations that are reported in the original study, rather than a sub-set of these mutations.

2) There are a number of problems with the original experimental design that complicate conclusions drawn from the xenograft study. Chroscinski and colleagues should be aware that: a) the genetic status of PREX2 in the cell line that is employed is unknown; b) the relative expression of endogneous PREX2 and ectopically expressed PREX2 is unknown; and c) it has previously been reported that WT PREX2 in a different cells (MCF10A) causes PTEN inhibition, activation of AKT, and increased proliferation (Fine et al. (2009) Science 325, 1261). These deficiencies in the design of the original study will influence the ability to draw sound conclusions from the study, but will be common between the original study and the replication study.

3) Regarding the power calculations, the proposed power calculations take for granted survival and hazard numbers published in the original study. This is fine at this stage, however, we suggest the following improvements.

(a) Cross-study variation should be taken into account to determine expected loos of power computed on published numbers, pre-data collection. This is hard to estimate, but papers by Giovanni Parmigiani and collaborators at the Dana Farber provide some estimates about cross-study variation that could be used for this purpose. The authors should budget some additional variability because of cross-study reproducibility, and increase the sample size on-the-fly, as they deem appropriate as deemed appropriate.

(b) The final report on the replicated study should report the actual power of the tests, based on the standard deviations in the replicated study.

https://doi.org/10.7554/eLife.04180.002

Author response

There are two main conclusions drawn in the Nature paper by Berger et al. First, PREX2 is identified as a frequently (14%) non-synonymously mutated gene in melanoma. Second, that ectopic expression of cancer-associated PREX2 proteins promote melanoma genesis in a xenograft assay in mice. This is an important study because the identification of driver (vs passenger) mutations in cancer is critical for understanding the mechanism of tumorigenesis.

The first conclusion is addressed by Chroscinski and colleagues in the literature summary. The authors note that validation of this conclusion is not supported by several studies, including meta-analysis. However, Chroscinski and colleagues state that two papers support the conclusion that PREX2 is frequently mutated in melanoma. This is not correct. Turajlic et al. do describe a PREX2 mutation, but the analysis is limited to a single patient, while Furney et al. only refer to PREX2 mutations described by Berger et al. Thus, these papers do not support the conclusion that PREX2 is frequently mutated in melanoma. It also appears to be significant that another study of melanoma exome sequencing by many of the same authors published 2 months after Berger et al (Hodis et al.) does not appear to identify PREX2 as a frequently mutated gene. Together, these considerations do not provide support for the conclusion presented by Berger et al. that PREX2 is frequently mutated in melanoma. This needs to be more directly addressed by Chroscinski and colleagues: they should discuss statistical thresholds for calling mutations significant (e.g., MutSig) and examine PREX2 mutations in public cancer genome portals such as cBioPortal (MSK) or the Broad Tumor Portal algorithms (in addition to reporting on the negative studies).

We thank the reviewers for these astute suggestions. We have amended the Introduction to more accurately describe the findings of Turajlic et al., and we have removed the incorrectly used reference for Furney et al. We have added a sentence to highlight the possibility of false-positive findings due to tumor heterogeneity, as described by Lawrence, et al. We have also replaced the phrase “significantly mutated” with “frequently mutated” in the Introduction.

In general, the Reproducibility Project: Cancer Biology focuses on generation of new data, not reanalysis of existing datasets. As such, while interesting, we feel it is beyond the scope of this replication study to perform data mining or analyses of PREX2 mutations in various genomic databases to determine prevalence or significance. Rather, we will instead limit our focus to replicating experiments that were performed in the original paper.

The second conclusion (that is addressed by re-analysis by Chroscinski and colleagues) is important: that PREX2 is a driver mutation rather than a passenger mutation during melanoma formation. The proposed experimental design appears to replicate the original study. However, a number of issues were raised by the reviewers:

1) The PREX2 mutations reported by Berger et al. are present throughout the PREX2 sequence. This is more consistent with passenger mutation than driver mutation. The restriction of the re-analysis to a limited number of mutations originally examined is therefore problematic. To document that the PREX2 mutations acts as drivers, it would be best to examine the same mutations that are reported in the original study, rather than a sub-set of these mutations.

We thank the reviewers for this insightful comment. However, ultimately, the goal of the Reproducibility Project: Cancer Biology is not to appraise the biological conclusions and implications imparted by the original authors, but rather to systematically assess the degree to which we can reproduce the methodology and experimental effect sizes described in the original paper. Of note, the original authors also chose to only analyze a subset (6/28) of all of the PREX2 mutations they identified (see Figure 3A).

We agree that all of the experiments included in the original study are important, and choosing which experiments to replicate has been one of the great challenges of this project. We acknowledge that the exclusion of certain experiments limits the scope of what can be analyzed about the project, but we are attempting to identify a balance of breadth of sampling for general inference with sensible investment of resources on replication projects.

Consistent with this mentality, we feel it is beyond the scope of this replication study to assess the original authors’ conclusions and interpretations regarding whether PREX2 is a passenger or driver in melanoma. We believe that replicating a subset of the original data is appropriate to allow us achieve our goal of quantitatively evaluating the ability of the originally reported results to be replicated. To avoid confusion regarding this issue, we have altered the Introduction so that it does not reference the debate over passenger versus driver mutations. We will also refrain from discussing the functional relevance of any of the mutations that we are not directly testing, including those identified but not analyzed in the original paper.

2) There are a number of problems with the original experimental design that complicate conclusions drawn from the xenograft study. Chroscinski and colleagues should be aware that: a) the genetic status of PREX2 in the cell line that is employed is unknown; b) the relative expression of endogneous PREX2 and ectopically expressed PREX2 is unknown; and c) it has previously been reported that WT PREX2 in a different cells (MCF10A) causes PTEN inhibition, activation of AKT, and increased proliferation (Fine et al. (2009) Science 325, 1261). These deficiencies in the design of the original study will influence the ability to draw sound conclusions from the study, but will be common between the original study and the replication study.

We thank the reviewers for these suggestions. We remind the reviewers that this project focuses on direct replication of the experiments as detailed in the original report and with information provided by the original authors. Aspects of an experiment not included in the original study are occasionally added to ensure the quality of the research, but by no means is a requirement of this project; rather, it is an extension of the original work. Adding additional aspects not included in the original study can be of scientific interest, and can be included if it is possible to balance them with the main aim of this project: to perform a direct replication of the original experiment(s).

As such, we agree with the reviewers that there is scientific interest in better understanding some aspects of this biological system. To address the reviewers’ concern (a) about the genetic status of endogenous PREX2, we have added an additional step to Protocol 1, whereby the endogenous PREX2 gene in NRASG12D melanocytes will be sequenced to determine its mutational status. Additionally, to address the reviewers’ concern (b) about the expression levels of endogenous PREX2, we have added an additional step to Protocol 2, whereby we will also blot for PREX2 protein in both the overexpressed PREX2 variants and the GFP vector control, comparing the levels of PREX2 expression across cell lines. However, we will consider these data exploratory and therefore not include them in our statistical analysis. As for the reviewers’ concern (c) regarding the behavior of PREX2 in different cell lines, we believe addressing this concern is beyond the scope of this particular replication study.

3) Regarding the power calculations, the proposed power calculations take for granted survival and hazard numbers published in the original study. This is fine at this stage, however, we suggest the following improvements.

(a) Cross-study variation should be taken into account to determine expected loos of power computed on published numbers, pre-data collection. This is hard to estimate, but papers by Giovanni Parmigiani and collaborators at the Dana Farber provide some estimates about cross-study variation that could be used for this purpose. The authors should budget some additional variability because of cross-study reproducibility, and increase the sample size on-the-fly, as they deem appropriate as deemed appropriate.

We thank the reviewers for these suggestions. The cross-study variation, such as approaches that utilize the 95% confidence interval of the effect size, can be useful in conducting power calculations when planning adequate sample sizes for detecting the true population effect size, which requires a range of possible observed effect sizes. However, the Reproducibility Project: Cancer Biology is designed to conduct replications that have 80% power to detect the point estimate of the originally reported effect size. While this has the limitation of being underpowered to detect smaller effects than what is originally reported, this standardizes the approach across all studies to be designed to detect the originally reported effect size with at least 80% power. Also, while the minimum power guarantee is beneficial for observing a range of possible effect sizes, the experiments in this replication, and all experiments in the project, are designed to detect the originally reported effect size with a minimum power of 80%. Thus, performing power calculations during or after data collection is not necessary in this replication attempt as all studies included are already designed to meet a minimum power or are identified beforehand as being underpowered and thus are not included in the confirmatory analysis plan. The papers by Giovanni Parmigiani and collaborators highlight the importance of accounting for variability that can occur across different studies, specifically gene expression data. While it is possible for a difference in variance between the originally reported results and the replication data, this will be reflected in the presentation of the data and a possible reason for obtaining a different effect size estimate.

(b) The final report on the replicated study should report the actual power of the tests, based on the standard deviations in the replicated study.

As described above, we do not see the value in performing post-hoc power calculations on the obtained data. However, we do agree that reporting the actual power of the tests to detect the originally reported effect size estimate based on the sample size analyzed in the replication study is important and will be reported.

https://doi.org/10.7554/eLife.04180.003

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Denise Chroscinski
  2. Darryl Sampey
  3. Alex Hewitt
  4. Reproducibility Project: Cancer Biology
(2014)
Registered report: Melanoma genome sequencing reveals frequent PREX2 mutations
eLife 3:e04180.
https://doi.org/10.7554/eLife.04180

Share this article

https://doi.org/10.7554/eLife.04180