Single-cell profiling reveals the intratumor heterogeneity and immunosuppressive microenvironment in cervical adenocarcinoma

  1. Fourth Department of Gynecologic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha 410013, Hunan, China
  2. Department of Pathology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha 410013, Hunan, China
  3. Department of Gynecologic Oncology, Changsha Kexin Cancer Hospital, 292 Fenglinsan Road, Changsha 410004, Hunan, China
  4. Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
  5. Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China
  6. Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China
  7. Department of Pathology, Third Hospital, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, public reviews, and a provisional response from the authors.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Lynne-Marie Postovit
    Queens University, Kingston, Canada
  • Senior Editor
    Lynne-Marie Postovit
    Queens University, Kingston, Canada

Reviewer #1 (Public Review):

Summary:

The authors in this manuscript performed scRNA-seq on a cohort of 15 early-stage cervical cancer patients with a mixture of adeno- and squamous cell carcinoma, HPV status, and several samples that were upstaged at the time of surgery. From their analyses they identified differential cell populations in both immune and tumour subsets related to stage, HPV status, and whether a sample was adenocarcinoma or squamous cell. Putative microenvironmental signaling was explored as a potential explanation for their differential cell populations. Through these analyses the authors also identified SLC26A3 as a potential biomarker for later stage/lymph node metastasis which was verified by IHC and IF. The dataset is likely useful for the community, however, the strong claims made are not adequately supported by the data and would require additional functional validation.

Strengths:

The dataset could be useful for the community.
SLC26A3 could potentially be a useful marker to predict lymph node metastasis with further study.

Weaknesses:

The link between the background in the introduction and the actual study and findings is often tenuous or not clearly explained. A re-working of the intro to better set up and link to the study questions would be beneficial.

For the sequencing, which kit was used on the Novaseq6000?

Additional details are needed for the analysis pipeline. How were batch effects identified/dealt with, what were the precise functions and settings for each step of the analysis, how was clustering performed and how were clusters validated etc. Currently, all that is given is software and sometimes function names which are entirely inadequate to be able to assess the validity of the analysis pipeline. This could alternatively be answered by providing annotated copies of the scripts used for analysis as a supplement.

For Cell type annotation, please provide the complete list of "selected gene markers" that were used for annotation.

No statistics are given for the claims on cell proportion differences throughout the paper (for cell types early, epithelial sub-clusters later, and immune cell subsets further on). This should be a multivariate analysis to account for ADC/SCC, HPV+/- and Early/Late stage.

The Y-axis label is missing from the proportion histograms in Figure 2D. In these same panels, the bars change widths on the right side. If these are exclusively in ADC, show it with a 0 bar for SCC, not doubling the width which visually makes them appear more important by taking up more area on the plot.

Throughout the manuscript, informatic predictions (differentiation potential, malignancy score, stemness, and trajectory) are presented as though they're concrete facts rather than the predictions they are. Strong conclusions are drawn on the basis of these predictions which do not have adequate data to support. These conclusions which touch on essentially all of the major claims made in the manuscript would need functional data to validate, or the claims need to be very substantially softened as they lack concrete support. Indeed, the fact that most of the genes examined that were characteristic of a given cluster did not show the expected expression patterns in IHC highlights the fact that such predictions require validation to be able to draw proper inferences.

The cluster Epi_10_CYSTM1 which is the basis for much of the paper is present in a single individual (with a single cell coming from another person), and heavily unconnected from the rest of the epithelial populations. If so much emphasis is placed on it, the existence of this cluster as a true subset of cells requires validation.

Claims based on survival analysis of TCGA for Epi_10_CYSTM1 are based on a non-significant p-value, though there is a slight trend in that direction.

The claim "The identification of Epi_10_CYSTM1 as the only cell cluster found in patients with stage IIICp raises the possibility that this cluster may be a potential marker to diagnose patients with lymph node metastasis." This is incorrect according to the sample distributions which clearly show cells from the patient who has EPI_10_CYSTM1 in multiple other clusters. This is then used as justification for SLC26A3 which appears to be associated with associated with late stage, however, in the images SLC26A3 appears to be broadly expressed in later tumours rather than restricted to a minor subset as it should be if it were actually related to the EPI_10_CYSTM1 cluster.

The authors claim that cytotoxic T cells express KRT17, and KRT19. This likely represents a mis-clustering of epithelial cells.

Multiple claims are made for specific activities based on GO term biological process analysis which while not contradictory to the data, certainly are by no means the only explanation for it, nor directly supported.

Reviewer #2 (Public Review):

Summary:

Peng et al. present a study using scRNA-seq to examine phenotypic properties of cervical cancer, contrasting features of both adenocarcinomas (ADC) and squamous cell carcinoma (SCC), and HPV-positive and negative tumours. They propose several key findings: unique malignant phenotypes in ADC with elevated stemness and aggressive features, interactions of these populations with immune cells to promote an immunosuppressive TME, and SLC26A3 as a biomarker for metastatic (>=Stage III ) tumours.

Strengths:

This study provides a valuable resource of scRNA-seq data from a well-curated collection of patient samples. The analysis provides a high-level view of the cellular composition of cervical cancers. The authors introduce some mechanistic explanations of immunosuppression and the involvement of regulatory T cells that are intriguing.

Weaknesses:

I believe that many of the proposed conclusions are over-interpretations or unwarranted generalizations of the single-cell analysis. These conclusions are often based on populations in the scRNA-seq data that are described as enriched or specific to a given group of samples (eg. ADC). This conclusion is based on the percentage of cells in that population belonging to the given group; for example, a cluster of cells that dominantly come from ADC. The data includes multiple samples for each group, but statistical approaches are never used to demonstrate the reproducibility of these claims.

This leads to problematic conclusions. For example, the "ADC-specific" Epi_10_CYSTM1 cluster, which is a central focus of the paper, only contains cells from one of the 11 ADC samples and represents only a small fraction of the malignant cells from that sample (Sample 7, Figure 2A). Yet, this population is used to derive SLC26A3 as a potential biomarker. SLC26A3 transcripts were only detected in this small population of cells (none of the other ADC samples), which makes me question the specificity of the IHC staining on the validation cohort.

This is compounded by technical aspects of the analysis that hinder interpretation. For example, it is clear that the clustering does not perfectly segregate cell types. In Figures 2B and D, it is evident that C4 and C5 contain mixtures of cell type (eg. half of C4 is EPCAM+/CD3-, the other half EPCAM-/CD3+). These contaminations are carried forward into subclustering and are not addressed. Rather, it is claimed that there is a T cell population that is CD3- and EPCAM+, which does not seem likely.

Author response:

Reviewer #1 (Public review):

(1) The link between the background in the introduction and the actual study and findings is often tenuous or not clearly explained. A re-working of the intro to better set up and link to the study questions would be beneficial.

Response: upon revision, we plan to rewrite the introduction of the manuscript.

(2) For the sequencing, which kit was used on the Novaseq6000?

Response: for sequencing, we used the Chromium Controller and Chromium Single Cell 3’Reagent Kits (v3 chemistry CG000183) on the Novaseq6000. We feel sorry for lacking this quite important part and will add the information in Methods.

(3) Additional details are needed for the analysis pipeline. How were batch effects identified/dealt with, what were the precise functions and settings for each step of the analysis, how was clustering performed and how were clusters validated etc. Currently, all that is given is software and sometimes function names which are entirely inadequate to be able to assess the validity of the analysis pipeline. This could alternatively be answered by providing annotated copies of the scripts used for analysis as a supplement.

Response: we apologize for the inadequacy of descriptions of data analysis process due to word count limit. We plan to provide more information, and if possible we also would like to provide scripts as supplementary data in the revised manuscript.

(4) For Cell type annotation, please provide the complete list of "selected gene markers" that were used for annotation.

Response: we will add the list of marker genes for cell type annotation in the revised manuscript.

(5) No statistics are given for the claims on cell proportion differences throughout the paper (for cell types early, epithelial sub-clusters later, and immune cell subsets further on). This should be a multivariate analysis to account for ADC/SCC, HPV+/- and Early/Late stage.

Response: considering this inadequacy, we plan to use statistic approaches for further analyses to compare the differences between each set of groups up revision.

(6) The Y-axis label is missing from the proportion histograms in Figure 2D. In these same panels, the bars change widths on the right side. If these are exclusively in ADC, show it with a 0 bar for SCC, not doubling the width which visually makes them appear more important by taking up more area on the plot.

Response: we feel sorry for impreciseness when presenting histograms such as Fig 2D and we will add labels in Y-axis. As for the width of bars, we just used the histograms generated originally from the data package. However, we did not intend to double the width on purpose to strengthen the visual importance. We sincerely feel sorry for this and will correct the similar mistakes alongside the whole manuscript.

(7) Throughout the manuscript, informatic predictions (differentiation potential, malignancy score, stemness, and trajectory) are presented as though they're concrete facts rather than the predictions they are. Strong conclusions are drawn on the basis of these predictions which do not have adequate data to support. These conclusions which touch on essentially all of the major claims made in the manuscript would need functional data to validate, or the claims need to be very substantially softened as they lack concrete support. Indeed, the fact that most of the genes examined that were characteristic of a given cluster did not show the expected expression patterns in IHC highlights the fact that such predictions require validation to be able to draw proper inferences.

Response: we agree that many conclusions, which were based on bio-informatic predictions, are written in an over-affirmative way. Upon revision, we will rewrite these conclusions more precisely.

(8) The cluster Epi_10_CYSTM1 which is the basis for much of the paper is present in a single individual (with a single cell coming from another person), and heavily unconnected from the rest of the epithelial populations. If so much emphasis is placed on it, the existence of this cluster as a true subset of cells requires validation.

Response: we are thankful for this suggestion. We think that each cluster of epithelial cells is specified from other clusters and identified by DEGs, but they are not heavily unconnected from others. Upon revision, we plan to add further validation for the existence of Epi_10_CYSTM1.

(9) Claims based on survival analysis of TCGA for Epi_10_CYSTM1 are based on a non-significant p-value, though there is a slight trend in that direction.

Response: from the data of TCGA survival analysis for Epi_10, we found a not-so-slight trend of difference between groups (with a small P value). As a result, we presented this data and hoped to add more strength to the clinical significance of this cluster. However, this indeed caused controversy because the P value is non-significant. We plan to rewrite the conclusion more precisely or delete this data in the revised manuscript.

(10) The claim "The identification of Epi_10_CYSTM1 as the only cell cluster found in patients with stage IIICp raises the possibility that this cluster may be a potential marker to diagnose patients with lymph node metastasis." This is incorrect according to the sample distributions which clearly show cells from the patient who has EPI_10_CYSTM1 in multiple other clusters. This is then used as justification for SLC26A3 which appears to be associated with associated with late stage, however, in the images SLC26A3 appears to be broadly expressed in later tumours rather than restricted to a minor subset as it should be if it were actually related to the EPI_10_CYSTM1 cluster.

Response: we feel thankful for this question. The conclusion “The identification of Epi_10_CYSTM1 as the only cell cluster found in patients with stage IIICp raises the possibility that this cluster may be a potential marker to diagnose patients with lymph node metastasis” has indeed been written too concrete according to the sample distribution. We will correct the description in the up-coming revised manuscript. As for SLC26A3, we also do not think it is “broadly” expressed, but it is specified in later tumors. When we presented the data of IHC, we only showed the strongly-positive area of each slide in order to emphasize the differences, however, this has caused misunderstandings. Thus, upon revision, we would like to show the other areas of one case or even the scan of one whole slide as supplementary data.

(11) The authors claim that cytotoxic T cells express KRT17, and KRT19. This likely represents a mis-clustering of epithelial cells.

Response: we apologize for the ignorance of further validation of cytotoxic T cells. From fig. 4B and 4C, the four different clusters of T cells were basically identified based on canonical T cell markers. And then we focused mainly on the validation and further analysis of Tregs, neglecting the other clusters. In fig. 4D we intended to only show the top DEGs in each T cell cluster and hoped to find some potential marker genes for next-step analysis. However, we did not notice that there might be contamination of epithelial cells within cytotoxic T cells when clustering. We will optimize the analysis of this part in our revision.

(12) Multiple claims are made for specific activities based on GO term biological process analysis which while not contradictory to the data, certainly are by no means the only explanation for it, nor directly supported.

Response: our initial purpose was to use GO analysis as supports for our conclusions. However we know these are only claims but not evidence, which is also the problem of our writing techniques as in question (7). Therefore, in our revised manuscript, we plan to rewrite the conclusion from the GO analysis in a more scientific way or delete these data.

Reviewer #2 (Public review):

(1) I believe that many of the proposed conclusions are over-interpretations or unwarranted generalizations of the single-cell analysis. These conclusions are often based on populations in the scRNA-seq data that are described as enriched or specific to a given group of samples (eg. ADC). This conclusion is based on the percentage of cells in that population belonging to the given group; for example, a cluster of cells that dominantly come from ADC. The data includes multiple samples for each group, but statistical approaches are never used to demonstrate the reproducibility of these claims.

Response: we understand that many of the conclusions are too sure but lack profound supporting evidence, thus we will optimize the writing in the revised manuscript. More importantly, to strengthen the validity of our data, we will try to use statistical approaches for further analysis.

(2) This leads to problematic conclusions. For example, the "ADC-specific" Epi_10_CYSTM1 cluster, which is a central focus of the paper, only contains cells from one of the 11 ADC samples and represents only a small fraction of the malignant cells from that sample (Sample 7, Figure 2A). Yet, this population is used to derive SLC26A3 as a potential biomarker. SLC26A3 transcripts were only detected in this small population of cells (none of the other ADC samples), which makes me question the specificity of the IHC staining on the validation cohort.

Response: we sincerely feel grateful for being questioned on the validity, appropriateness and the real potential of SLC26A3. We plan to add more explanation of the importance of SLC26A3 in the discussion part. We are also sorry for some over-sure conclusions about ADC-specific cell clusters, as well as the marker gene SLC26A3. However, we do not think these conclusions are problematic. In fact, due to the heterogeneity among different individuals, as well as even different sites within one individual when sampling, we think a “small faction” does not means it will not make sense. Also, these ADC-specific clusters (including Epi_10_CYSTM1) do have certain proportions when comparing with those “big fraction” groups (Fig. 2D). Furthermore, when considering the specificity of DEGs to ADC only, but not to SCC, we think it might be these ADC-specific cluster genes to have the central function to make a difference between ADC and SCC. And we further used validation experiment to support our hypothesis. Lastly and most importantly, SLC26A3 was coming from sample 7 whose clinical stage is FIGO IIIC (late stage) and pathological type is ADC. Among the 15 cases, there are only 4 cases whose clinical stages are late (within which 3 are ADC). At this point of view, we think 1 in 3 (33%) having expression of SLC26A3 (or existence of cluster Epi_10_CYSTM1) should be considered as a potential choice. Samples coming from early-staged and SCC patients do not have fractions of Epi_10_CYSTM1. This likewise indicates the specificity of this cell cluster to ADC. Therefore, in our revised manuscript, we plan to add more in-depth discussion about this question.

(3) This is compounded by technical aspects of the analysis that hinder interpretation. For example, it is clear that the clustering does not perfectly segregate cell types. In Figures 2B and D, it is evident that C4 and C5 contain mixtures of cell type (eg. half of C4 is EPCAM+/CD3-, the other half EPCAM-/CD3+). These contaminations are carried forward into subclustering and are not addressed. Rather, it is claimed that there is a T cell population that is CD3- and EPCAM+, which does not seem likely.

Response: do you mean Figure 1B and D? In the revised manuscript, we will list the canonical marker genes to cluster different types of cells to at least support that the clustering of cell types match most of the present published references. To further avoid the contamination of cells in each cluster, we will use quality controls and re-analyze these data upon revision.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation