Exposure-Response Relationships in Patients With HER2-Positive Metastatic Breast Cancer and Other Solid Tumors Treated With Trastuzumab Deruxtecan

Ophelia Yin, Hiroji Iwata, Chia-Chi Lin, Kenji Tamura, Junichiro Watanabe, Russ Wada, Helen Kastrissios, Malaz AbuTarif, Tushar Garimella, Caleb Lee, Lin Zhang, Javad Shahidi, Frank LaCreta, Ophelia Yin, Hiroji Iwata, Chia-Chi Lin, Kenji Tamura, Junichiro Watanabe, Russ Wada, Helen Kastrissios, Malaz AbuTarif, Tushar Garimella, Caleb Lee, Lin Zhang, Javad Shahidi, Frank LaCreta

Abstract

Trastuzumab deruxtecan (T-DXd) is a HER2-targeting antibody-drug conjugate composed of a novel enzyme-cleavable linker and membrane-permeable topoisomerase I inhibitor payload. T-DXd has been approved for HER2-positive metastatic breast cancer and for HER2-positive metastatic gastric cancer. The approval in breast cancer was based on results from the DESTINY-Breast01 (U201; NCT03248492) and J101 (NCT02564900) trials. Here, we present dose justification for the approved 5.4 mg/kg every-3-weeks (Q3W) dose based on exposure-efficacy evaluated in patients with HER2-positive breast cancer (N = 337) from these 2 trials. Exposure-safety was assessed in patients with all tumor types (N = 639, n = 512 with breast cancer) across 5 trials, including J101 and DESTINY-Breast01. T-DXd doses ranged from 0.8-8.0 mg/kg Q3W; most patients received 5.4 (n = 312) or 6.4 mg/kg (n = 291). For each end point, multivariate logistic or Cox regression analysis was performed using various exposure metrics of T-DXd and released drug. A statistically significant association was observed between intact T-DXd area under the concentration-time curve (AUC) and confirmed objective response rate (ORR; P = 0.028). No significant exposure-response relationships were observed between intact T-DXd or released drug and duration of response or progression-free survival; however, follow-up was limited. All evaluated safety end points demonstrated a significant (P < 0.05) relationship with either intact T-DXd or released drug, with higher adverse event (AE) rates projected at higher exposures. Dose-response projections suggested an increase in ORR (67.5% vs. 62.9%) and toxicity (e.g., grade ≥ 3 all-cause treatment-emergent AEs: 61% vs. 54%) with T-DXd 6.4 vs. 5.4 mg/kg. Results demonstrate the benefit-risk profile at different doses and guide clinicians in the use of the 5.4-mg/kg Q3W dose in patients with HER2-positive metastatic breast cancer.

Conflict of interest statement

O.Y., T.G., M.A.T., L.Z., J.S., and F.L. report employment with and equity ownership in Daiichi Sankyo. C.L. reports employment with Daiichi Sankyo. H.I. reports honoraria from AstraZeneca, Chugai/Roche, Daiichi Sankyo, Eisai, Lilly, Novartis, Pfizer, and Taiho, and consulting fees from AbbVie, AstraZeneca, Chugai/Roche, Daiichi Sankyo, Kyowa Hakko Kirin, Lilly, Novartis, Odonate, and Pfizer. C.‐C.L. reports travel support from BeiGene and Daiichi Sankyo, advisory fees from Blueprint Medicines, Boehringer Ingelheim, Daiichi Sankyo, and Novartis, and honoraria from Boehringer Ingelheim, Lilly, Novartis, and Roche. K.T. reports receiving research funding from Daiichi Sankyo, Eisai, Lilly, and Pfizer. J.W. reports personal fees from Daiichi Sankyo related to the current work and lecture fees from AstraZeneca, Chugai, Eisai, Eli Lilly, Novartis, and Pfizer outside of the submitted work. R.W. and H.K. report consulting fees from Daiichi Sankyo related to the current work and consulting fees from undisclosed sponsors outside of the submitted work.

© 2021 The Authors. Clinical Pharmacology & Therapeutics published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Exposure‐response plot (a) and forest plot of confirmed central ORR by Cavg (b) and exposure‐response plot of confirmed central ORR by Cmin (c). In panels a and c, numbers in red represent n/N; horizontal bars below the plot illustrate the 5th, 50th, and 95th percentiles of exposures. Cavg, average concentration; Cmin, minimum concentration; ORR, objective response rate.
Figure 2
Figure 2
Kaplan‐Meier curves of duration of response (a) and progression‐free survival (b) stratified by intact T‐DXd exposure. Cavg, average concentration; ORR, objective response rate; PFS, progression‐free survival; Q, quartile; T‐DXd, trastuzumab deruxtecan.
Figure 3
Figure 3
Logistic regression plots of discontinuations associated with AEs (a) and anemia (b) and Cox regression plots of ILD (c). In panels a and b, numbers in red represent n/N; horizontal bars below the plot illustrate the 5th, 50th, and 95th percentiles of exposures. In panel c, the solid black line represents the estimated Kaplan‐Meier curves, the dashed lines are 95% CIs, and the solid green line represents the estimated Kaplan‐Meier curve determined using multivariate Cox regression. AE, adverse event; AUC, area under the concentration‐time curve; Cavg, average concentration; CI, confidence interval; Cmax, maximum concentration; ILD, interstitial lung disease; Q, quartile.
Figure 4
Figure 4
Forest plots of probability of discontinuations associated with AEs (a), anemia (b), and ILD (c). AE, adverse event; AUCSS, area under the concentration‐time curve at steady state; Cavg, average concentration; ECOG PS, Eastern Cooperative Oncology Group performance status; ILD, interstitial lung disease.
Figure 5
Figure 5
Model‐projected event rates for T‐DXd 5.4‐ and 6.4‐mg/kg doses in the overall population (all patients), non‐Asian group, Asian‐Japan group, and Asian–non‐Japan group. Error bars represent 90% CI. No effect of race‐country was observed on ORR; therefore, the ORR on all race‐country plots represents the ORR for the overall population. Discontinuations, dose interruptions, and dose reductions plotted are those due to adverse events. Anemia, neutropenia, and thrombocytopenia are laboratory test based. ECHO, echocardiogram; ILD; interstitial lung disease; LVEF, left ventricular ejection fraction; ORR, objective response rate; SAE, serious adverse event.

References

    1. Doi, T. et al. Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS‐8201), a HER2‐targeting antibody‐drug conjugate, in patients with advanced breast and gastric or gastro‐oesophageal tumours: a phase 1 dose‐escalation study. Lancet Oncol. 18, 1512–1522 (2017).
    1. Aggarwal, N. & Sloane, B.F. Cathepsin B: multiple roles in cancer. Proteomics Clin. Appl. 8, 427–437 (2014).
    1. Nakada, T. , Sugihara, K. , Jikoh, T. , Abe, Y. & Agatsuma, T. The latest research and development into the antibody‐drug conjugate, [fam‐] trastuzumab deruxtecan (DS‐8201a), for HER2 cancer therapy. Chem. Pharm. Bull. (Tokyo) 67, 173–185 (2019).
    1. Ruan, J. , Zheng, H. , Fu, W. , Zhao, P. , Su, N. & Luo, R. Increased expression of cathepsin L: a novel independent prognostic marker of worse outcome in hepatocellular carcinoma patients. PLoS One 9, e112136 (2014).
    1. Ogitani, Y. et al. DS‐8201a, a novel HER2‐targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T‐DM1. Clin. Cancer Res. 22, 5097–5108 (2016).
    1. Enhertu (fam‐trastuzumab deruxtecan‐nxki) [package insert]. Basking Ridge, NJ: Daiichi Sankyo, Inc; 2020.
    1. Tamura, K. et al. Trastuzumab deruxtecan (DS‐8201a) in patients with advanced HER2‐positive breast cancer previously treated with trastuzumab emtansine: a dose‐expansion, phase 1 study. Lancet Oncol. 20, 816–826 (2019).
    1. Yin, O. et al. Population pharmacokinetic analysis of DS‐8201a ([Fam‐] trastuzumab deruxtecan), a HER2‐targeting antibody‐drug conjugate, in patients with HER2‐positive breast cancer or other solid tumors. Clin. Pharmacol. Ther. 105(suppl 1) (2019). 10.1002/cpt.1344
    1. Center for Drug Evaluation and Research . Multi‐discipline review: Enhertu <> (2019). Accessed April 20, 2021.
    1. Modi, S. et al. Trastuzumab deruxtecan in previously treated HER2‐positive breast cancer. N. Engl. J. Med. 382, 610–621 (2020).
    1. Tamura, K. et al. Dose justification for DS‐8201a, a HER2‐targeted antibody‐drug conjugate, for HER2‐positive breast cancer: observed clinical data and exposure‐response analyses. Cancer Res. 79(4 suppl), abstract P6–17‐10 (2019). 10.1158/1538-7445.SABCS18-P6-17-10
    1. Yin, O. et al. Population pharmacokinetics of trastuzumab deruxtecan in patients with HER2‐positive breast cancer and other solid tumors. Clin. Pharmacol. Ther. 109, 1314–1325 (2021).
    1. Chang, D.‐Y. et al. Safety and pharmacokinetic results from a phase 1, multicenter, open‐label study of [fam‐] trastuzumab deruxtecan (T‐DXd; DS‐8201a) in subjects with advanced HER2‐positive breast cancer. Mol. Cancer Ther. 18, abstract C041 (2019).
    1. Center for Drug Evaluation and Research . Clinical pharmacology and biopharmaceutics review: Kadcyla <> (2012). Accessed January 17, 2020.
    1. Bajaj, G. , Wang, X. , Agrawal, S. , Gupta, M. , Roy, A. & Feng, Y. Model‐based population pharmacokinetic analysis of nivolumab in patients with solid tumors. CPT Pharmacometrics Syst. Pharmacol. 6, 58–66 (2017).
    1. Wang, Y. , Booth, B. , Rahman, A. , Kim, G. , Huang, S.M. & Zineh, I. Toward greater insights on pharmacokinetics and exposure‐response relationships for therapeutic biologics in oncology drug development. Clin. Pharmacol. Ther. 101, 582–584 (2017).
    1. Ait‐Oudhia, S. , Zhang, W. & Mager, D.E. A mechanism‐based PK/PD model for hematological toxicities induced by antibody‐drug conjugates. AAPS J. 19, 1436–1448 (2017).
    1. Thon, J.N. , Devine, M.T. , Jurak Begonja, A. , Tibbitts, J. & Italiano, J.E. Jr. High‐content live‐cell imaging assay used to establish mechanism of trastuzumab emtansine (T‐DM1)–mediated inhibition of platelet production. Blood 120, 1975–1984 (2012).
    1. Uppal, H. et al. Potential mechanisms for thrombocytopenia development with trastuzumab emtansine (T‐DM1). Clin. Cancer Res. 21, 123–133 (2015).
    1. Herceptin (trastuzumab) [package insert]. South San Francisco, CA: Genentech, Inc; 2017.
    1. Kadcyla (ado‐trastuzumab emtansine) [package insert]. South San Francisco, CA: Genentech, Inc; 2016.
    1. Dimopoulou, I. , Bamias, A. , Lyberopoulos, P. & Dimopoulos, M.A. Pulmonary toxicity from novel antineoplastic agents. Ann. Oncol. 17, 372–379 (2006).
    1. Ewer, S.M. & Ewer, M.S. Cardiotoxicity profile of trastuzumab. Drug Saf. 31, 459–467 (2008).
    1. Shitara, K. et al. Trastuzumab deruxtecan (DS‐8201a) in patients with advanced HER2‐positive gastric cancer: a dose‐expansion, phase 1 study. Lancet Oncol. 20, 827–836 (2019).
    1. Azuma, A. & Kudoh, S. High prevalence of drug‐induced pneumonia in Japan. Japan Med. Assoc. J. 50, 405–411 (2007).
    1. Kudoh, S. et al. Interstitial lung disease in Japanese patients with lung cancer. Am. J. Respir. Crit. Care Med. 177, 1348–1357 (2008).
    1. Namba, N. et al. Association of MUC5B promoter polymorphism with interstitial lung disease in myeloperoxidase‐antineutrophil cytoplasmic antibody‐associated vasculitis. Ann. Rheum. Dis. 78, 1144–1146 (2019).

Source: PubMed

3
订阅