Targeting Ribosome Biogenesis as a Novel Therapeutic Approach to Overcome EMT-related Chemoresistance in Breast Cancer

  1. Department of Cardiothoracic Surgery
  2. Sandra and Edward Meyer Cancer Center Weill Cornell Medicine, 1300 York Avenue, New York, New York 10065
  3. Neuberger Berman Lung Cancer Center
  4. Systems Medicine and Bioengineering Department Houston Methodist Cancer Center, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX 77030
  5. Department of Radiology
  6. Department of Pathology and Laboratory Medicine
  7. Department of Cell and Developmental Biology

Peer review process

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

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Yongliang Yang
    Dalian University of Technology, Dalian, China
  • Senior Editor
    Caigang Liu
    Shengjing Hospital of China Medical University, Shenyang, China

Reviewer #1 (Public Review):

The process of EMT is a major contributor to metastasis and chemoresistance in breast cancer. By using a modified PyMT model that allows the identification of cells undergoing EMT and their decedents via S100A4-Cre mediated recombination of the mTmG allele, Ban et al. tackle a very important question of how tumor metastasis and therapy resistance by EMT can be blocked. They identified that pathways associated with ribosome biogenesis (RiBi) are activated during transition cell states. This finding represents a promising therapeutic target to block any transition from E to M (activated during cell dissemination and invasion) as well as from M to E (activated during metastatic colonization). Inhibition of RiBi-blocked EMT also reduced the establishment of chemoresistance that is associated with an EMT phenotype. Hence, RiBi blockage together with standard chemotherapy showed synergistic effects, resulting in impaired colonization/metastatic outgrowth in an animal model. The study is of great interest and of high clinical relevance as the authors show that blocking the transition from E to M or vice versa targets both aspects of metastasis, dissemination from the primary tumor, and colonization in distant organs.

The study is done with high skill using state-of-the-art technology and the conclusions are convincing and solid, but some aspects require some additional experimental support and clarification. It remains elusive whether blocking of EMT/MET is necessary for the synergistic effect of standard chemotherapy together with RiBi blockage or whether a general growth disadvantage of RiBi-treated cells independent of blocking transition is responsible. How can specific effects on state transition by RiBI block be separated from global effects attributed to overall reduced protein biosynthesis, proliferation etc.? Some other aspects are misleading or need extension.

Reviewer #2 (Public Review):

The current manuscript by Ban et al describes that cells undergoing EMT have increased rRNA synthesis, as analyzed by RNA seq-based gene expression analysis, and that the increased rRNA synthesis provides a therapeutic opportunity to target chemoresistance. The cells utilized in this manuscript were isolated from the authors' Tri-PyMT EMT lineage tracing model published a few years ago which demonstrated that cells undergoing EMT are not the cells that are contributing to metastasis but rather to tumor chemoresistance (Fischer, Nature 2015). This in vivo model has since then been criticized for not capturing all relevant EMT events which the authors also acknowledge in the introduction. The authors therefore reason that they use this lineage tracing model to better understand the role of EMT in chemoresistance.

A major problem with the current manuscript is that the authors present many of their findings as a novel without the proper acknowledgment of previously published literature in particular, Prakash et al., Nature Communications, 2019 and Dermitt, Dev Cell, 2020. In the studies by Prakash, the authors demonstrate that maintaining ongoing rRNA biogenesis is essential for the execution of the EMT program, and thus the ability of cancer cells to become migratory and invasive. Further, Prakash et al showed that blocking rRNA biogenesis with a small molecule inhibitor, CX-5461 (which is also used in the study by Ban et al) specifically inhibits breast cancer growth, invasion, EMT, and metastasis in animal models without significant toxicity to normal tissues. As such a significant revision that is necessary at this time is a rewrite of the manuscript especially the introduction and the discussion to more accurately describe and cite previously published findings and then highlight the current work by Ban et al which nicely builds on the previously published literature as it highlights the contribution of EMT to chemoresistance rather than metastasis. The suggestion for the authors is that they therefore should focus on highlighting the chemotherapy resistance angle as their Tri-PyMT EMT lineage tracing was chosen to test this angle and as such focus on both primary tumor growth and metastasis.

Additional major revisions:
The authors use the FSP1-Cre Model which in the field has been questioned as to not capture all the relevant EMT events and therefore their findings should be corroborated by another EMT model system.

In the current version of the manuscript, there are no measurements of rRNA synthesis, but the gene expression profiles are used as a proxy for rRNA synthesis. The authors therefore need to include measurements of rRNA synthesis corroborating the RNA sequencing data to support their scientific findings and claims. This can be accomplished by qPCR, Northern blot, or EU staining of the respective sorted cell population. Quantification of rRNA synthesis is also needed for the CX-5461/BMH-21 and silencing studies.

Currently, there is no mechanistic insight as to how rRNA synthesis is increased during EMT, which would also strengthen the manuscript. This could be done through targeted ChIP analysis.

rRNA synthesis has canonically been linked to the cell cycle therefore it will be necessary for the authors to determine the cell cycle state of their respective cell populations throughout the manuscript.

Statistics and quantifications are currently missing in several figures and need to be better explained throughout the manuscript to strengthen the scientific rigor of the studies.

Only metastasis studies are shown in the current version of the manuscript. These studies should be complemented with primary tumor studies as the main focus of the paper is the contribution of EMT to chemoresistance.

Reviewer #3 (Public Review):

Summary:
Ban et al. investigated the role of ribosome biogenesis (RiBi) in epithelial-to-mesenchymal transition (EMT) and its contribution to chemoresistance in breast cancer. They used a Tri-PyMT EMT lineage-tracing model and scRNA-seq to analyze EMT status and found that RiBi was elevated during both EMT and mesenchymal-to-epithelial transition (MET) of cancer cells. They further revealed that nascent protein synthesis mediated by ERK and mTOR signaling pathways was essential for the completion of RiBi. Inhibiting excessive RiBi impaired EMT and MET capability. More importantly, combinatorial treatment with RiBi inhibitors and chemotherapy drugs reduced metastatic outgrowth of both epithelial and mesenchymal tumor cells. These results suggest that targeting the RiBi pathway may be an effective strategy for treating advanced breast cancer with EMT-related chemoresistance.

Strengths:
The conclusions of this study are generally supported by the data. However, some weaknesses still exist as mentioned below.

Weaknesses:

  1. The study predominantly focused on RiBi as a target for overcoming EMT-related chemoresistance. Thus, it will be necessary to provide some canonical outcomes after upregulating ribosome biogenesis, such as translation activity. I would suggest ribosome profiling or puromycin-incorporation assay, or other more suitable experiments.

  2. The results were basically obtained from mice and in vitro experiments. While these results provide valuable insights, it will be valuable to validate part of the findings using some tissue samples from patients (e.g. RiBi activity) to determine the clinical relevance and potential therapeutic applications.

  3. The results revealed that mTORC1 and ERK mediated RiBi activation. How about mTORC2? It will be informative to evaluate mTORC2 signaling.

  4. The results also demonstrated promising synergic effects of Pol I inhibitor (BMH21) and chemotherapy drug (CTX) on chemo-resistant metastasis. How about using the inhibitors of mTORC1 together with CTX?

  5. While the results demonstrate the potential efficacy of RiBi inhibitors in reducing metastatic outgrowth, other factors and mechanisms contributing to chemoresistance may exist and need further investigation. I would suggest some discussion about this aspect.

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