Inhibitor-induced HER2-HER3 heterodimerisation promotes proliferation through a novel dimer interface

  1. Jeroen Claus
  2. Gargi Patel
  3. Flavia Autore
  4. Audrey Colomba
  5. Gregory Weitsman
  6. Tanya N Soliman
  7. Selene Roberts
  8. Laura C Zanetti-Domingues
  9. Michael Hirsch
  10. Francesca Collu
  11. Roger George
  12. Elena Ortiz-Zapater
  13. Paul R Barber
  14. Boris Vojnovic
  15. Yosef Yarden
  16. Marisa L Martin-Fernandez
  17. Angus Cameron  Is a corresponding author
  18. Franca Fraternali  Is a corresponding author
  19. Tony Ng  Is a corresponding author
  20. Peter J Parker  Is a corresponding author
  1. The Francis Crick Institute, United Kingdom
  2. Kings College London, United Kingdom
  3. Brighton and Sussex University Hospitals, United States
  4. Rutherford Appleton Laboratory, United Kingdom
  5. King’s College London, Guy’s Hospital, United Kingdom
  6. University College London, United Kingdom
  7. Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, United Kingdom
  8. Weizmann Institute of Science, Israel
  9. Queen Mary University of London, United Kingdom
  10. Guy’s Hospital King’s College London School of Medicine, United Kingdom
  11. King’s College London, Guy’s Campus, United Kingdom

Decision letter

  1. Benjamin F Cravatt
    Reviewing Editor; The Scripps Research Institute, United States

In the interests of transparency, eLife includes the editorial decision letter and accompanying author responses. A lightly edited version of the letter sent to the authors after peer review is shown, indicating the most substantive concerns; minor comments are not usually included.

Thank you for submitting your article "Inhibitor-induced HER2-HER3 heterodimerisation promotes proliferation through a novel dimer interface" for consideration by eLife. Your article has been reviewed by two peer reviewers, and the evaluation has been overseen by a Reviewing Editor and Jonathan Cooper as the Senior Editor. The following individual involved in review of your submission has agreed to reveal his identity: Nathanael S Gray (Reviewer #2).

The reviewers have discussed the reviews with one another and the Reviewing Editor has drafted this decision to help you prepare a revised submission.

The reviewers were generally quite impressed by the manuscript, noting that it represented a very nice molecular pharmacology detective story to try to explain the paradoxical activation of Her2 signaling upon treatment with an ATP-competitive Her2 inhibitor Lapatinib. Key observations supported by the data presented include 1) the Her2/Her3 dimer induced by lapatinib + neuregulin does not resemble the head to tail model for "normal" activation by neuregulin alone, but a head to head orientation, previously only observed in the HER3 crystal structure 2) the Her3 ATP site needs to be occupied by a ligand (ATP or bosutinib) for the hyperactivation to occur, 3) an irreversible version of lapatinib (neratinib) does not activate cell growth – presumably because the compound is irreversibly bound to HER2. The reviewers felt that these observations are an important contribution to our understanding of HER2/HER3 signaling in breast cancer. However, several points were raised for revision, including some that will require additional experiments.

Essential revisions:

1) The authors use various mutations in Her3 to show ATP binding by Her3 is important to maintain activating ability of lapatinib. They should show CETSA or other binding data for what these mutations do to bosutinib affinity of Her3 in cells. Also I wondered what bosutinib combined with lapatinib does in the presence of these mutations (synergy or antagonism and do the mutations abrogate this effect)? The authors could also have used the covalent Her3 binder TX1-85-1 (Nat Chem Biol. 2014 Dec; 10(12): 1006-1012.) which is more selective than bosutinib as a way to interrogate what Her3 ATP-site ligation does to the pharmacology of lapatinib.

2) Much of the mechanistic analysis is done in SKBR3 cells, which from the data presented appear to be somewhat of an outlier in terms of the Neuregulin + lapatinib induction of growth. In Figure 1—figure supplement 1 the authors investigate three other cells, BT474, MCF7, and ZR75. The first is a high Her2 expresser and also shows the same Neuregulin + lapatinib enhanced growth. Several more HER2 overexpressing cell lines should be tested for this effect (2-5 should suffice).

https://doi.org/10.7554/eLife.32271.036

Author response

Essential revisions:

1) The authors use various mutations in Her3 to show ATP binding by Her3 is important to maintain activating ability of lapatinib. They should show CETSA or other binding data for what these mutations do to bosutinib affinity of Her3 in cells. Also I wondered what bosutinib combined with lapatinib does in the presence of these mutations (synergy or antagonism and do the mutations abrogate this effect)? The authors could also have used the covalent Her3 binder TX1-85-1 (Nat Chem Biol. 2014 Dec; 10(12): 1006-1012.) which is more selective than bosutinib as a way to interrogate what Her3 ATP-site ligation does to the pharmacology of lapatinib.

Based on the reviewer’s suggestions we have included CETSA binding data of COS7 cells transfected with HER3wt, HER3KGG or the bosutinib-desensitised mutation HER3T787M. We compared the recovery of HER3 protein between 44°C and 50°C in the presence or absence of bosutinib and show that bosutinib stabilises HER3wt, but it does not stabilise either the HER3KGG or HER3T787M mutations. This corresponds to our proliferation data to suggest that HER3KGG does not have strong affinity for bosutinib in cells. This data is included in the revised manuscript as Figure 2—figure supplement 1G.

The reviewers suggest the use of the covalent HER3 inhibitor TX1-85-1 as a more selective inhibitor than bosutinib. In the original publication cited (Xie et al., 2014) the authors demonstrate TX1-85-1 potently inhibits HER2 (Supplementary Table 1 in Xie et al.). Although the covalent binding to HER3 would make TX1-85-1 an interesting tool to probe ATP-binding pocket behaviour in HER3, the fact that it is also a potent HER2 binder would confound the analysis in the proposed assays.

HER3 has been reported to be the only EGFR family member that tightly binds bosutinib due to an alanine equivalent to SrcA403, which is lacking in other EGFR family members (Levinson et al. Nat Chem Biol 2013, DOI 10.1038/NCHEMBIO.1404). This is supported by screening data from Davis et al., 2011. To verify, we have included in vitro TSA analysis of HER2 kinase domain treated with Mg2+/ATP, lapatinib or bosutinib. This data agrees with the published results on HER2-bosutinib binding, showing that HER2 is not a strong binder of bosutinib in our hands. This data is included in the revised manuscript as Figure 2B.

2) Much of the mechanistic analysis is done in SKBR3 cells, which from the data presented appear to be somewhat of an outlier in terms of the Neuregulin + lapatinib induction of growth. In Figure 1—figure supplement 1 the authors investigate three other cells, BT474, MCF7, and ZR75. The first is a high Her2 expresser and also shows the same Neuregulin + lapatinib enhanced growth. Several more HER2 overexpressing cell lines should be tested for this effect (2-5 should suffice).

As the reviewers suggested, we have included additional cell lines in the revised manuscript and reclassified the presentation of the various cell lines in Figure 1—figure supplement 1. Instead of referring to the HER2 expressions status, we present the data as ‘lapatinib-responding’, ‘partial lapatinib response’ and ‘lapatinib non-responding’, based on the cellular toxicity presented by lapatinib. We match the phenotype seen in SKBR3 with BT474 and AU565, show a partial match with ZR75 and HCC1419, and a negative response with MCF7 and HCC1569.

https://doi.org/10.7554/eLife.32271.037

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. Jeroen Claus
  2. Gargi Patel
  3. Flavia Autore
  4. Audrey Colomba
  5. Gregory Weitsman
  6. Tanya N Soliman
  7. Selene Roberts
  8. Laura C Zanetti-Domingues
  9. Michael Hirsch
  10. Francesca Collu
  11. Roger George
  12. Elena Ortiz-Zapater
  13. Paul R Barber
  14. Boris Vojnovic
  15. Yosef Yarden
  16. Marisa L Martin-Fernandez
  17. Angus Cameron
  18. Franca Fraternali
  19. Tony Ng
  20. Peter J Parker
(2018)
Inhibitor-induced HER2-HER3 heterodimerisation promotes proliferation through a novel dimer interface
eLife 7:e32271.
https://doi.org/10.7554/eLife.32271

Share this article

https://doi.org/10.7554/eLife.32271