Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

HER kinase inhibition in patients with HER2- and HER3-mutant cancers

An Author Correction to this article was published on 12 February 2019

Abstract

Somatic mutations of ERBB2 and ERBB3 (which encode HER2 and HER3, respectively) are found in a wide range of cancers. Preclinical modelling suggests that a subset of these mutations lead to constitutive HER2 activation, but most remain biologically uncharacterized. Here we define the biological and therapeutic importance of known oncogenic HER2 and HER3 mutations and variants of unknown biological importance by conducting a multi-histology, genomically selected, ‘basket’ trial using the pan-HER kinase inhibitor neratinib (SUMMIT; clinicaltrials.gov identifier NCT01953926). Efficacy in HER2-mutant cancers varied as a function of both tumour type and mutant allele to a degree not predicted by preclinical models, with the greatest activity seen in breast, cervical and biliary cancers and with tumours that contain kinase domain missense mutations. This study demonstrates how a molecularly driven clinical trial can be used to refine our biological understanding of both characterized and new genomic alterations with potential broad applicability for advancing the paradigm of genome-driven oncology.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Individual treatment outcome and response for 141 patients grouped by tumour cohort and mutant allele/domain.
Figure 2: Integrated efficacy by tumour type and HER2 allele/domain.
Figure 3: Genomic modifiers of response and outcome by treatment duration.

Similar content being viewed by others

References

  1. Chmielecki, J. et al. Oncogenic alterations in ERBB2/HER2 represent potential therapeutic targets across tumors from diverse anatomic sites of origin. Oncologist 20, 7–12 (2015)

    Article  CAS  PubMed  Google Scholar 

  2. Zehir, A. et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 23, 703–713 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Schram, A. et al. Landscape of somatic ERBB2 mutations: Findings from AACR GENIE and comparison to ongoing ERBB2 mutant basket study. Cancer Res. 77, Abstract LB-103 (2017)

    Google Scholar 

  4. Chang, M. T. et al. Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity. Nat. Biotechnol. 34, 155–163 (2016)

    Article  CAS  PubMed  Google Scholar 

  5. Bose, R. et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 3, 224–237 (2013)

    Article  CAS  PubMed  Google Scholar 

  6. Kavuri, S. M. et al. HER2 activating mutations are targets for colorectal cancer treatment. Cancer Discov. 5, 832–841 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jaiswal, B. S. et al. Oncogenic ERBB3 mutations in human cancers. Cancer Cell 23, 603–617 (2013)

    Article  CAS  PubMed  Google Scholar 

  8. Chumsri, S. et al. Prolonged response to trastuzumab in a patient with HER2-nonamplified breast cancer with elevated HER2 dimerization harboring an ERBB2 S310F mutation. J. Natl. Compr. Canc. Netw. 13, 1066–1070 (2015)

    Article  CAS  PubMed  Google Scholar 

  9. Zabransky, D. J. et al. HER2 missense mutations have distinct effects on oncogenic signaling and migration. Proc. Natl Acad. Sci. USA 112, E6205–E6214 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hyman, D. M. et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N. Engl. J. Med. 373, 726–736 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hyman, D. M. et al. AKT inhibition in solid tumors with AKT1 mutations. J. Clin. Oncol. 35, 2251–2259 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ross, J. S. et al. A high frequency of activating extracellular domain ERBB2 (HER2) mutation in micropapillary urothelial carcinoma. Clin. Cancer Res. 20, 68–75 (2014)

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Ross, J. S. et al. Relapsed classic E-cadherin (CDH1)-mutated invasive lobular breast cancer shows a high frequency of HER2 (ERBB2) gene mutations. Clin. Cancer Res. 19, 2668–2676 (2013)

    Article  CAS  PubMed  Google Scholar 

  14. Wolff, A. C. et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J. Clin. Oncol. 31, 3997–4013 (2013)

    Article  PubMed  Google Scholar 

  15. Ma, C. X. et al. Neratinib efficacy and circulating tumor DNA detection of HER2 mutations in HER2 non-amplified metastatic breast cancer. Clin. Cancer Res. 23, 5687–5695 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yasuda, H. et al. Structural, biochemical, and clinical characterization of epidermal growth factor receptor (EGFR) exon 20 insertion mutations in lung cancer. Sci. Transl. Med. 5, 216ra177 (2013)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Borghaei, H. et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kosaka, T. et al. Response heterogeneity of EGFR and HER2 exon 20 insertions to covalent EGFR and HER2 inhibitors. Cancer Res. 77, 2712–2721 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Freedman, R. A. et al. Translational Breast Cancer Research Consortium (TBCRC) 022: a phase II trial of neratinib for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases. J. Clin. Oncol. 34, 945–952 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Shi, W. et al. Pathway level alterations rather than mutations in single genes predict response to HER2-targeted therapies in the neo-ALTTO trial. Ann. Oncol. 28, 128–135 (2017)

    CAS  PubMed  Google Scholar 

  21. Loibl, S. et al. PIK3CA mutations are associated with reduced pathological complete response rates in primary HER2-positive breast cancer: pooled analysis of 967 patients from five prospective trials investigating lapatinib and trastuzumab. Ann. Oncol. 27, 1519–1525 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Baselga, J. et al. Biomarker analyses in CLEOPATRA: a phase III, placebo-controlled study of pertuzumab in human epidermal growth factor receptor 2-positive, first-line metastatic breast cancer. J. Clin. Oncol. 32, 3753–3761 (2014)

    Article  CAS  PubMed  Google Scholar 

  23. Schram, A. M., Berger, M. F. & Hyman, D. M. Precision oncology: charting a path forward to broader deployment of genomic profiling. PLoS Med. 14, e1002242 (2017)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Hyman, D. M., Taylor, B. S. & Baselga, J. Implementing genome-driven oncology. Cell 168, 584–599 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jordan, E. J. et al. Prospective comprehensive molecular characterization of lung adenocarcinomas for efficient patient matching to approved and emerging therapies. Cancer Discov. 7, 596–609 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Baselga, J. et al. Phase II study of weekly intravenous recombinant humanized anti-p185HER2 monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer. J. Clin. Oncol. 14, 737–744 (1996)

    Article  CAS  PubMed  Google Scholar 

  27. Vogel, C. L. et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J. Clin. Oncol. 20, 719–726 (2002)

    Article  CAS  PubMed  Google Scholar 

  28. Slamon, D. J. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001)

    Article  CAS  PubMed  Google Scholar 

  29. Bang, Y. J. et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376, 687–697 (2010)

    Article  CAS  PubMed  Google Scholar 

  30. Swain, S. M. et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N. Engl. J. Med. 372, 724–734 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Blackwell, K. L. et al. Randomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J. Clin. Oncol. 28, 1124–1130 (2010)

    Article  CAS  PubMed  Google Scholar 

  32. Baselga, J. et al. Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet 379, 633–640 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bertotti, A. et al. A molecularly annotated platform of patient-derived xenografts (‘xenopatients’) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 1, 508–523 (2011)

    Article  CAS  PubMed  Google Scholar 

  34. Sartore-Bianchi, A. et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 17, 738–746 (2016)

    Article  CAS  PubMed  Google Scholar 

  35. Kaufman, B. et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J. Clin. Oncol. 33, 244–250 (2015)

    Article  CAS  PubMed  Google Scholar 

  36. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hyman, D. M. et al. The efficacy of larotrectinib (LOXO-101), a selective tropomyosin receptor kinase (TRK) inhibitor, in adult and pediatric TRK fusion cancers. J. Clin. Oncol. 35, abstract LBA2501 (2017)

    Article  Google Scholar 

  38. Wahl, R. L., Jacene, H., Kasamon, Y. & Lodge, M. A. From RECIST to PERCIST: Evolving considerations for PET response criteria in solid tumors. J. Nucl. Med. 50, 122S–150S (2009)

    Article  CAS  PubMed  Google Scholar 

  39. Cheng, D. T. et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J. Mol. Diagn. 17, 251–264 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chang, M. T. et al. Accelerating discovery of functional mutant alleles in cancer. Cancer Discov. https://doi.org/10.1158/2159-8290.CD-17-0321 (2017)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Kanehisa, M. & Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28, 27–30 (2000)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chakravarty D. et al. OncoKB: a precision oncology knowledge base. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00011 (2017)

    Article  Google Scholar 

  43. Shen, R. & Seshan, V. E. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 44, e131 (2016)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Carter, S. L. et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 30, 413–421 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. McGranahan, N. et al. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl. Med. 7, 283ra54 (2015)

    Article  PubMed  PubMed Central  Google Scholar 

  46. Niu, B. et al. MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics 30, 1015–1016 (2014)

    Article  CAS  PubMed  Google Scholar 

  47. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Middha et al. Reliable pan-cancer microsatellite instability assessment by using targeted next-generation sequencing data. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00084 (2017)

  49. Zhou, X. et al. Exploring genomic alteration in pediatric cancer using ProteinPaint. Nat. Genet. 48, 4–6 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank patients and their families for participating in this study. Editorial support, not including writing, was provided by L. Miller. This work was funded by Puma Biotechnology, and supported by grants from the National Institutes of Health (grants P30 CA008748, P30 CA016672, P30 CA014089, R01 CA204749, R01 CA80195, T32 CA009207, 1U01 CA180964 and UL1 TR000371), the National Institutes of Health/National Cancer Institute (Breast SPORE grant P50 CA098131), Cycle for Survival, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, The Cancer Prevention and Research Institute of Texas (RP1100584), the Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, Nellie B. Connally Breast Cancer Research Endowment, and the Breast Cancer Research Foundation.

Author information

Authors and Affiliations

Authors

Contributions

D.M.H., H.W., M.F.B., R.E.C, F.X., A.B., L.D.E., G.M., C.F., A.S.L., R.P.B., J.B. and D.B.S. designed the study and supervised the analyses. R.E.C., F.X., L.D.E., G.M., C.F., A.S.L. and R.P.B. helped to collect and monitor the clinical outcome data. D.M.H., S.A.P., J.R., C.S., G.I.S., D.J., D.I.Q., V.M., B.D., I.A.M., V.B., E.C., S.L., A.C.L., J.P.E., B.T.L., A.J.H., R.M., A.M.S., A.D., L.M.S., K.J., G.I., J.J.H., C.L.A., F.M.B., J.B. and D.B.D. enrolled patients and provided patient samples. G.U. developed the PET response criteria and performed radiographic response assessments. B.S.T., J.P., J.T., S.D.S., N.B., M.M., M.F.B., J.B. and D.B.S. performed the tumour and plasma sequencing, provided computational infrastructure, and made final variant calls. D.M.H., H.W., M.S., B.S.T., J.P., J.T., H.B., M.F.B. and D.B.S. analysed clinical and genomic data and performed the integrated efficacy analyses. F.X. performed biostatistical analyses of the clinical efficacy data. D.M.H., H.W., B.S.T., C.L.A., F.M.B. and D.B.S. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to David M. Hyman.

Ethics declarations

Competing interests

R.E.C., F.X., L.D.E., G.M., C.F., A.S.L. and R.P.B. are employees of Puma Biotechnology. D.M.H., M.S. and J.B. receive research support from Puma Biotechnology, B.T.L. and M.S. receive research funding from Diachi, A.D. receives personal fees from Roche, and D.S. received personal fees from Loxo Oncology and Pfizer.

Additional information

Reviewer Information Nature thanks E. Mardis and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Figure 1 Design of SUMMIT study.

Five tumour-specific HER2 (ERBB2)-mutant cohorts were pre-specified (endometrial, gastroesophageal, ovarian, colorectal and bladder/urinary tract). In addition, a sixth ‘solid tumour (not otherwise specified, NOS)’ HER2-mutant cohort allowed for the enrolment of patients with any other cancer types. A sufficient number of patients with breast, cervical, biliary and lung cancer were enrolled in the solid tumours (NOS) cohort to permit independent efficacy analysis using the same design as the pre-specified cohorts. Patients with HER3 (ERBB3)-mutant tumours were enrolled in a HER3-specific cohort regardless of tumour type. CBR, clinical benefit rate; cfDNA, cell-free (tumour) DNA; CI, confidence interval.

Extended Data Figure 2 Distribution of HER2 and HER3 mutations positioned by their amino acid coordinates across the respective protein domains.

a, b, HER2 (a) and HER3 (b) mutations (125 and 16 mutations, respectively). Each unique mutation is represented by a circle, with the circle size and number representing the frequency, and coloured to show the mutation class as indicated in the legend. The corresponding amino acid change and common hotspot mutations (shown in pink) are labelled next to the circles.

Source data

Extended Data Figure 3 Spectrum of HER2 and HER3 mutations observed in the neratinib study versus TCGA, ICGC and other public datasets.

a, b, Distribution of HER2 (a) and HER3 (b) mutations observed across our cohort in comparison to the spectrum of HER2 and HER3 mutations (reflected lollipop) from publically available datasets (TCGA, ICGC and other published studies).

Source data

Extended Data Figure 4 Distribution and outcome of 28 HER2 exon 20 insertions.

a, Percentage best change and PFS plots corresponding to each type of exon 20 insertion (colour coded by synonymous amino acid change). Three cases with no change are indicated in colour-coded circles above the x axis. b, Zoomed-in schematic of all exon 20 insertions positioned by their amino acid coordinates and frequencies. c, Five unique types of exon 20 insertions observed in the study with the resulting full amino acid sequences (insertion indicated in red).

Source data

Extended Data Figure 5 Genomic modifiers of response and outcome by treatment duration.

a, Cancer cell fractions with 95% confidence intervals and clonality status of all HER2 mutations in 74 patients with sufficient sequencing data ordered by increasing clinical benefit (weeks on therapy). b, Comparison of the percentage activation of known oncogenic alterations in the three pathways between the patients of clinical benefit (n = 20, biologically independent samples) and no benefit (n = 66, biologically independent samples). Nominal Fisher’s P values are shown.

Source data

Extended Data Figure 6 SUMMIT CONSORT diagram.

Extended Data Table 1 Patient demographics and efficacy by cohort
Extended Data Table 2 Treatment-emergent adverse events (occurring in ≥ 10% of patients)
Extended Data Table 3 PET response criteria
Extended Data Table 4 Patient disposition by cohort

Supplementary information

Life Sciences Reporting Summary (PDF 72 kb)

Supplementary Information

This file contains: 1 - list of genes covered in the MSK-IMPACT panel along with the HGNC ID, short gene description, chromosomal location, and panel version, 2 - list of all somatic mutations within the MSK-IMPACT genes for patient tumour samples with sequencing data and 3 - list of all somatic copy number alterations within the MSK-IMPACT genes for patient tumour samples with sequencing data. (PDF 745 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hyman, D., Piha-Paul, S., Won, H. et al. HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature 554, 189–194 (2018). https://doi.org/10.1038/nature25475

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature25475

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing