Ado-trastuzumab emtansine (T-DM1) in patients with HER2-amplified tumors excluding breast and gastric/gastroesophageal junction (GEJ) adenocarcinomas: results from the NCI-MATCH trial (EAY131) subprotocol Q - PubMed (original) (raw)

Clinical Trial

. 2019 Nov 1;30(11):1821-1830.

doi: 10.1093/annonc/mdz291.

X V Wang 2, V Makker 3, S-W Luoh 4, E P Mitchell 5, J A Zwiebel 6, E Sharon 7, R J Gray 8, S Li 8, L M McShane 9, L V Rubinstein 10, D Patton 11, P M Williams 12, S R Hamilton 13, B A Conley 14, C L Arteaga 15, L N Harris 14, P J O'Dwyer 16, A P Chen 17, K T Flaherty 18

Affiliations

Clinical Trial

Ado-trastuzumab emtansine (T-DM1) in patients with HER2-amplified tumors excluding breast and gastric/gastroesophageal junction (GEJ) adenocarcinomas: results from the NCI-MATCH trial (EAY131) subprotocol Q

K L Jhaveri et al. Ann Oncol. 2019.

Erratum in

Abstract

Background: The National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) is a national precision medicine study incorporating centralized genomic testing to direct refractory cancer patients to molecularly targeted treatment subprotocols. This treatment subprotocol was designed to screen for potential signals of efficacy of ado-trastuzumab emtansine (T-DM1) in HER2-amplified histologies other than breast and gastroesophageal tumors.

Methods: Eligible patients had HER2 amplification at a copy number (CN) >7 based on targeted next-generation sequencing (NGS) with a custom Oncomine AmpliSeq™ (ThermoFisher Scientific) panel. Patients with prior trastuzumab, pertuzumab or T-DM1 treatment were excluded. Patients received T-DM1 at 3.6 mg/kg i.v. every 3 weeks until toxicity or disease progression. Tumor assessments occurred every three cycles. The primary end point was centrally assessed objective response rate (ORR). Exploratory end points included correlating response with HER2 CN by NGS. The impact of co-occurring genomic alterations and PTEN loss by immunohistochemistry were also assessed.

Results: Thirty-eight patients were enrolled and 36 included in efficacy analysis. Median prior therapies in the metastatic setting was 3 (range 0-9; unknown in one patient). Median HER2 CN was 17 (range 7-139). Partial responses were observed in two (5.6%) patients: one mucoepidermoid carcinoma of parotid gland and one parotid gland squamous cell cancer. Seventeen patients (47%) had stable disease including 8/10 (80%) with ovarian and uterine carcinomas, with median duration of 4.6 months. The 6-month progression-free survival rate was 23.6% [90% confidence interval 14.2% to 39.2%]. Common toxicities included fatigue, anemia, fever and thrombocytopenia with no new safety signals. There was a trend for tumor shrinkage with higher levels of gene CN as determined by the NGS assay.

Conclusion: T-DM1 was well tolerated. While this subprotocol did not meet the primary end point for ORR in this heavily pre-treated diverse patient population, clinical activity was seen in salivary gland tumors warranting further study in this tumor type in dedicated trials.

Keywords: HER2-amplified; NCI-MATCH; NGS; T-DM1.

© The Author(s) 2019. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.

PubMed Disclaimer

Figures

Figure 1.

Figure 1.

ERBB2 amplification frequency: all tumors excluding breast and gastric/gastroesophageal junction tumors. This plot includes solid tumors (467/28 106 samples and 442/25 637 patients) with a minimum of 50 sequenced tumors and ERBB2 amplification > 0% using MSK-IMPACT assay. The numbers in the parentheses are the total number of sequenced tumors. (cBioPortal.org).

Figure 2.

Figure 2.

Consort diagram.

Figure 3.

Figure 3.

(A) Efficacy: Best% change from baseline (n = 28). Four patients had PD based on new lesions but data for the target lesions are not available so not included in this plot. (B) Treatment Duration in patients who achieved SD or PR. (C) Kaplan–Meier curve for progression-free survival. GYN, gynecological; CI, confidence interval; GI, gastrointestinal.

Figure 4.

Figure 4.

Correlation of best % change by ERBB2 copy number gain (n = 28). PR, partial response; SD, stable disease; PD, progressive disease.

Figure 5.

Figure 5.

Co-occurring genomic alterations with HER2 amplification using the NCI-MATCH assay color coded by variant type (left): copy number variant (purple), single nucleotide variant (red), indel (pink) and number of variants (top) for each eligible patient along with information regarding histology and best response on treatment (bottom): partial response (PR), stable disease (SD), progression of disease (PD), unevaluable (UE).

Similar articles

Cited by

References

    1. Slamon DJ, Clark GM, Wong SG. et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987; 235(4785): 177–182. - PubMed
    1. Slamon DJ, Godolphin W, Jones LA. et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 1989; 244(4905): 707–712. - PubMed
    1. Wolff AC, Hammond ME, Hicks DG. 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. JCO 2013; 31(31): 3997–4013. - PubMed
    1. Bang YJ, Van Cutsem E, Feyereislova A. 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 2010; 376(9742): 687–697. - PubMed
    1. Slamon DJ, Leyland-Jones B, Shak S. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 2001; 344(11): 783–792. - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources