Computational Engineering of a Therapeutic Antibody to Inhibit Multiple Mutants of HER2 Without Compromising Inhibition of the Canonical HER2

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    In this important manuscript, the authors used unbiased approaches to identify somatic mutations in publicly available databases that would disrupt clinically approved antibodies targeting HER2. Using a solid combination of both computational and experimental approaches they identify mutations that could restore therapeutic antibody sensitivity in a series of disease-relevant model systems. Additional cell-based and in vivo assays would strengthen the work and increase the translational and potential clinical relevance of the proposed work.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

Genomic germline and somatic variations may impact drug binding and even lead to resistance. However, designing a different drug for each mutant may not be feasible. In this study, we identified the most common cancer somatic mutations from the Catalogue of Somatic Mutations in Cancer (COSMIC) that occur in structurally characterized binding sites of approved therapeutic antibodies. We found two HER2 mutations, S310Y and S310F, that substantially compromise binding of Pertuzumab, a widely used therapeutics, and lead to drug resistance. To address these mutations, we designed a multi-specific version of Pertuzumab, that retains original function while also bindings these HER2 variants. This new antibody is stable and inhibits HER3 phosphorylation in a cell-based assay for all three variants, suggesting it can inhibit HER2-HER3 dimerization in patients with any of the variants. This study demonstrates how a small number of carefully selected mutations can add new specificities to an existing antibody without compromising its original function, creating a single therapeutic antibody that targets multiple common variants, making a drug that is not personalized yet its activity may be.

Article activity feed

  1. Author Response

    Reviewer #1 (Public Review):

    Strengths:

    1. In my assessment, the data sufficiently demonstrates that a modified version of Pertuzamab can bind both the wild-type and S310 mutant forms of ERBB2.
    1. The engineering strategy employed is rational and effectively combines computational and experimental techniques.
    1. Given the clinical activity of HER2-targeting ADCs, antibodies unaffected by ERBB2 mutations would be desired.

    Weaknesses:

    1. There is no data showing that the engineered antibody is equally specific as Pertuzamab i.e. that it does not bind to other (non-ERBB2) proteins.

    Showing the specificity of the engineered antibodies is indeed important. We did not address it in the current ms, but it can be tested in the future.

    1. There is no data showing that the engineered antibody has the desired pharmacokinetics/pharmacodynamics properties or efficacy in vivo.

    In this ms we did not conduct in-vivo experiments. When moving forward, pharmacokinetics/pharmacodynamics properties and efficacy will be tested as well.

    1. Computational approaches are only used to design a phage-screen library, but not used to prioritize mutations that are likely to improve binding (e.g. based on predicted impact on the stability of the interaction). A demonstration of how computational pre-screening or lead optimization can improve the time-intensive process would be a welcome advance.

    Thank you for this important comment. In the present ms we indeed used a computational approach for prioritizing residues to be mutated, but we did not prioritize the mutations that are likely to improve binding. In the initial library design, we did prioritize the mutations. However, due to experimental approach limitations with codon’s selection for the library, we had decided to allow all possible residues in each position, knowing that the selection will remove non-binding variants.

    Context:

    The conflict of interest statement is inadequate. Most authors of the study (but not the first author) are employees of Biolojic, a company developing multi-specific antibodies, but the statements do not clarify whether the presented antibodies represent Biolojic IP, whether the company sponsored the research, and whether the company is further developing the specific antibodies presented.

    The Conflict-of-Interest statement will be revised as such: The Biolojic Design authors are employees of Biolojic Design and have stock options in Biolojic Design. The company did not sponsor the research, does not hold IP for the presented antibodies, and is not further developing the presented antibodies.

    Reviewer #2 (Public Review):

    Strengths:

    1. Deep computational analyses of large datasets of clinical data provide useful information about HER2 mutations and their potential relevance to antibody therapy resistance.
    1. There is valuable information analyzing the residues within or near the interface between the antigen HER2 and the Pertuzumab antibody (heavy chain). The experimental antibody library screening obtained 90+ clones from 3.86×1011 sequences for further functional validation.

    Weaknesses:

    1. There is a lack of assessment for antibody variant functions in cancer cell phenotypes in vitro (proliferation, cell death, motility) or in vivo (tumor growth and animal survival). The only assay was the western blotting of phosphopho-HER3 in Figure 4. However, HER2 levels and phosphor-HER2 were not analyzed.

    We indeed did not assess the engineered antibodies function in cancer cells. Regarding signaling assessment, previous works [1-3] also measured the signaling activation following HER2-HER3 dimerization by measuring pHER3, and we relied on them in this ms.

    1. There is a misleading impression from the title of computational engineering of a therapeutic antibody and the statement in the abstract "we designed a multi-specific version of Pertuzumab that retains original function while also bindings these HER2 variants" for a few reasons:

    a. The primary method used for variant antibody identification for HER2 mutant binding is rather traditional experimental screening based on yeast display instead of the computational design of a multi-specific version of Pertuzumab.

    b. There is insufficient or lack of computational power in the antibody design or prioritization in choosing variant residues for the library construction of 3.86×1011 sequences. It seems random combinations from 6 residues out of 4 groups with 20 amino acid options.

    c. The final version of the tri-binding variant is a combination of screened antibody clones instead of computation design from scratch.

    d. There is incomplete experimental evidence about the therapeutic values of newly obtained antibody clones.

    Thank you for this relevant comment. When addressing relevant residues to be mutated, the number of potential variants is enormous. The computational approach was aimed at identifying the most preferable residues, in which variation can improve binding and is not likely to harm important interactions. Although an initial smaller number of residues could be chosen, we decided to broaden our view and create a larger library, in the aim of combining the computational selection with an experimental selection. This indeed is not a computational design from scratch, but rather an intercourse between the computer and the lab, that yielded the presented results.

    1. Figures can be improved with better labeling and organization. Some essential pieces of data such as Supplementary Figure 1B on HER2 mutations in S310 that abrogated its binding to Pertuzumab should be placed in the main figures.

    Thank you for this comment, the relevant figures will be moved to the main text, and the labels will be revised.

    1. It is recommended to provide a clear rationale or flowchart overview into the main Figure 1. Figure 2A can be combined with Figure 1 to the list of targeted residues.

    Figures 1 and 2 will be divided differently, and the rationale will be detailed in the revised text.

    1. The quality of Figures such as Figure 2B-C flow data needs to be improved.

    This will be corrected in the revised text.

    1. Diwanji, D., et al., Structures of the HER2-HER3-NRG1β complex reveal a dynamic dimer interface. Nature, 2021. 600(7888): p. 339-343.

    2. Yamashita-Kashima, Y., et al., Mode of action of pertuzumab in combination with trastuzumab plus docetaxel therapy in a HER2-positive breast cancer xenograft model. Oncol Lett, 2017. 14(4): p. 4197-4205.

    3. Kang, J.C., et al., Engineering multivalent antibodies to target heregulin-induced HER3 signaling in breast cancer cells. MAbs, 2014. 6(2): p. 340-53.

  2. eLife assessment

    In this important manuscript, the authors used unbiased approaches to identify somatic mutations in publicly available databases that would disrupt clinically approved antibodies targeting HER2. Using a solid combination of both computational and experimental approaches they identify mutations that could restore therapeutic antibody sensitivity in a series of disease-relevant model systems. Additional cell-based and in vivo assays would strengthen the work and increase the translational and potential clinical relevance of the proposed work.

  3. Reviewer #1 (Public Review):

    Summary:
    Starting from an unbiased search for somatic mutations (from COSMIC) likely disrupting binding of clinically approved antibodies the authors focus on mutations known to disrupt binding between two ERBB2 mutations and Pertuzamab. They use a combined computational and experimental strategy to nominate a position that when mutated could result in restoring the therapeutic activity of the antibody. Using in vitro assays the authors confirm that the engineered antibody binds to the mutant ERBB2 and prevents ERBB3 phosphorylation

    Strengths:
    1. In my assessment, the data sufficiently demonstrates that a modified version of Pertuzamab can bind both the wild-type and S310 mutant forms of ERBB2.

    2. The engineering strategy employed is rational and effectively combines computational and experimental techniques.

    3. Given the clinical activity of HER2-targeting ADCs, antibodies unaffected by ERBB2 mutations would be desired.

    Weaknesses:
    1. There is no data showing that the engineered antibody is equally specific as Pertuzamab i.e. that it does not bind to other (non-ERBB2) proteins.

    2. There is no data showing that the engineered antibody has the desired pharmacokinetics/pharmacodynamics properties or efficacy in vivo.

    3. Computational approaches are only used to design a phage-screen library, but not used to prioritize mutations that are likely to improve binding (e.g. based on predicted impact on the stability of the interaction). A demonstration of how computational pre-screening or lead optimization can improve the time-intensive process would be a welcome advance.

    Context:
    The conflict of interest statement is inadequate. Most authors of the study (but not the first author) are employees of Biolojic, a company developing multi-specific antibodies, but the statements do not clarify whether the presented antibodies represent Biolojic IP, whether the company sponsored the research, and whether the company is further developing the specific antibodies presented.

  4. Reviewer #2 (Public Review):

    Summary:
    Peled et al identified HER2 mutations in connection with resistance to the anti-HER2 antibody Pertuzumab-mediated therapy. After constructing a yeast display library of Pertuzumab variants with 3.86×1011 sequences for targeted screening of variant combinations in chosen 6 out of 14 residues, the authors performed experimental screening to obtain the clones that bind to HER2 WT and/or mutants (S310Y and S310F), and then combined new variations to obtain antibodies with a broad spectrum binding to both WT and two HER2 mutants. These are interesting studies of clinical impact and translational potential.

    Strengths:
    1. Deep computational analyses of large datasets of clinical data provide useful information about HER2 mutations and their potential relevance to antibody therapy resistance.

    2. There is valuable information analyzing the residues within or near the interface between the antigen HER2 and the Pertuzumab antibody (heavy chain). The experimental antibody library screening obtained 90+ clones from 3.86×1011 sequences for further functional validation.

    Weaknesses:
    1. There is a lack of assessment for antibody variant functions in cancer cell phenotypes in vitro (proliferation, cell death, motility) or in vivo (tumor growth and animal survival). The only assay was the western blotting of phosphopho-HER3 in Figure 4. However, HER2 levels and phosphor-HER2 were not analyzed.

    2. There is a misleading impression from the title of computational engineering of a therapeutic antibody and the statement in the abstract "we designed a multi-specific version of Pertuzumab that retains original function while also bindings these HER2 variants" for a few reasons:
    a. The primary method used for variant antibody identification for HER2 mutant binding is rather traditional experimental screening based on yeast display instead of the computational design of a multi-specific version of Pertuzumab.
    b. There is insufficient or lack of computational power in the antibody design or prioritization in choosing variant residues for the library construction of 3.86×1011 sequences. It seems random combinations from 6 residues out of 4 groups with 20 amino acid options.
    c. The final version of the tri-binding variant is a combination of screened antibody clones instead of computation design from scratch.
    d. There is incomplete experimental evidence about the therapeutic values of newly obtained antibody clones.

    3. Figures can be improved with better labeling and organization. Some essential pieces of data such as Supplementary Figure 1B on HER2 mutations in S310 that abrogated its binding to Pertuzumab should be placed in the main figures.

    4. It is recommended to provide a clear rationale or flowchart overview into the main Figure 1. Figure 2A can be combined with Figure 1 to the list of targeted residues.

    5. The quality of Figures such as Figure 2B-C flow data needs to be improved.