ArticlePublisher preview availableLiterature Review

Exploring DESTINY: the Past, Present, and Future of Trastuzumab Deruxtecan

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Purpose of Review HER2-positive breast cancer accounts for 10–15% of all breast cancers and fam-trastuzumab deruxtecan (T-DXd) has played a major role in moving the treatment of HER2-expressing disease forward. Recent Findings T-DXd is a novel antibody–drug conjugate (ADC) composed of a humanized IgG1 monoclonal antibody against HER2 receptor bound to a potent topoisomerase I cytotoxin payload by a cleavable peptide linker. It has been shown to have robust preclinical activity in pretreated cancer cell lines, as well as meaningful clinical activity in advanced HER2-expressing breast cancer. Recent studies have demonstrated T-DXd as an active agent for metastatic HER2-positive patients, and as a viable additional line for heavily pretreated patients with HER2-low disease. The toxicity of T-DXd remains manageable and burden of side effects seems to be lower when offered as an earlier line of therapy over the course of treatment. Summary In this review, we discuss the pharmacology of T-DXd, review pertinent preclinical and clinical data, and address potential challenges and future directions related to the use of T-DXd in clinical practice.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
Current Oncology Reports (2024) 26:1–9
https://doi.org/10.1007/s11912-023-01478-2
Exploring DESTINY: thePast, Present, andFuture ofTrastuzumab
Deruxtecan
AzkaAli1 · StephanieL.Gra2
Accepted: 29 October 2023 / Published online: 13 December 2023
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2023
Abstract
Purpose of Review HER2-positive breast cancer accounts for 10–15% of all breast cancers and fam-trastuzumab deruxtecan
(T-DXd) has played a major role in moving the treatment of HER2-expressing disease forward.
Recent Findings T-DXd is a novel antibody–drug conjugate (ADC) composed of a humanized IgG1 monoclonal antibody
against HER2 receptor bound to a potent topoisomerase I cytotoxin payload by a cleavable peptide linker. It has been shown
to have robust preclinical activity in pretreated cancer cell lines, as well as meaningful clinical activity in advanced HER2-
expressing breast cancer. Recent studies have demonstrated T-DXd as an active agent for metastatic HER2-positive patients,
and as a viable additional line for heavily pretreated patients with HER2-low disease. The toxicity of T-DXd remains man-
ageable and burden of side effects seems to be lower when offered as an earlier line of therapy over the course of treatment.
Summary In this review, we discuss the pharmacology of T-DXd, review pertinent preclinical and clinical data, and address
potential challenges and future directions related to the use of T-DXd in clinical practice.
Keywords Breast cancer· HER2-positive· HER2-low· Metastatic breast cancer· Fam-traztuzumab deruxtecan, Antibody
drug conjugate· DESTINY Breast
Introduction
Human epidermal growth factor receptor-2 (HER2) directed
therapies have greatly improved the prognosis of HER2-
positive breast cancer in the early and metastatic settings
[1]. Several classes of anti-HER2 drugs have shown clini-
cal activity and been approved for the treatment of breast
cancer including anti-HER2 monoclonal antibodies (mAb),
HER2-targeted tyrosine kinase inhibitors, and antibody drug
conjugates (ADC) [1].
Fam-trastuzumab deruxtecan (T-DXd, or DS-8201a) is an
ADC, which is composed of a humanized immunoglobulin
G1 monoclonal antibody targeting HER2 followed by a
cleavable peptide linker bound to a derivative of DX-8951
(DXd), a potent topoisomerase I inhibitor payload [13].
Lysosomal enzymes such as cathepsins B and L cleave the
antibody and linker-payload, and anti-HER2 antibody binds
the HER2 receptor expressed on tumor cells, and membrane-
permeable payload is released after internalization of the
HER2-ADC complex [3].
Since the first introduction into phase I trials in 2015,
T-DXd has been studied as treatment of several HER2-
expressing cancers including HER2-positive and HER2-low
breast cancer, HER2-positive gastric cancer, HER2-express-
ing colorectal cancer, and HER2-expressing or mutated non-
small cell lung cancer [2].
In this review, we discuss development and unique phar-
macokinetics of T-DXd, review pertinent preclinical and
clinical data on T-DXd in breast cancer patients, address
pertinent adverse events (AEs) and management, and future
direction of research of T-DXd in relation to breast cancer.
* Azka Ali
alia22@ccf.og
Stephanie L. Graff
SGraff1@Lifespan.org
1 Department ofBreast Medical Oncology, Cleveland Clinic
Foundation, Taussig Cancer Institute, 9500 Euclid Ave /
CA-060, Cleveland, OH44195, USA
2 Division ofHematology & Medical Oncology, Lifespan
Cancer Institute, Legorreta Cancer Center atBrown
University, 593 Eddy St., Providence, RI02903, USA
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... 2,3 However, we have witnessed remarkable clinical progress in drug discovery for the treatment of individuals with diseases such as breast cancer, lung cancer, rheumatoid arthritis, and multiple sclerosis, which has raised hopes that we can do the same for AD and other related dementias. [8][9][10][11] Importantly, we note that acceptance of incremental increases in treatment efficacy, together with the concept of combination therapies, was instrumental to the successful pursuit of better therapeutic and clinical approaches for these and a spectrum of other conditions. However, the controversies that surround AD-modifying therapies may discourage similar therapeutic advancement unless recognized and addressed appropriately. ...
... To provide context for this important discussion, the current manuscript compares and contrasts perceptions on treatment with disease-modifying biologic agents across disparate conditions. 4,[8][9][10][11] The data presented include 2023 disease prevalence and mortality estimates for the United States and cost comparisons of five different biologic therapies used for multiple disparate and distinct disease states. The data provide important comparative insights into how the clinical meaningfulness and cost-effectiveness of these therapies are driving clinical decision-making today, as well as context for decision-making relevant to AD-oriented biologics. ...
Article
Full-text available
Introduction The advent of disease‐modifying therapies for Alzheimer's disease (AD) has raised many questions and debates in the field as to the clinical benefits, risks, and costs of such therapies. The controversies have resulted in the perception that many clinicians are apprehensive about prescribing these medications to their patient populations. There also remains widespread uncertainty as to the economic impact, cost benefit ratio, and safety oversight for use of these medications in standard clinical care settings. Methods To contextualize such issues, the present study compared anti‐amyloid biologic therapy (lecanemab) to four commonly used biologic agents in other fields, including trastuzumab for breast cancer, bevacizumab for lung cancer, etanercept for rheumatoid arthritis, and ocrelizumab for multiple sclerosis. Results The data presented demonstrate comparable costs, clinical benefits, and risks for these biologic agents in their disparate disease states. Discussion These results provide context for the costs, clinical benefits, and safety regarding the mainstream use of anti‐amyloid biologic agents for the prevention of cognitive loss. While the era of disease‐modifying therapies for AD is now in its infancy, there is an expectation that these discoveries will be followed by improved therapies and combination treatments leading to greater efficacy in ameliorating the clinical trajectory of AD. Highlights Anti‐amyloid therapy costs are comparable to other commonly used biologics. Anti‐amyloid therapy efficacy is comparable to other commonly used biologics. Anti‐amyloid therapy safety is compatible with other commonly used biologics.
... Although HER2 overexpression has long been considered necessary for the efficacy of anti-HER2 targeted therapy, new evidence suggests a change in treatment philosophy with the development of new and more effective agents such as T-DXd (Ali and Graff, 2024;Tarantino et al., 2023c). This ADC treatment involves a humanised HER2 antibody linked via a cleavable junction to a membrane-permeable payload of topoisomerase I inhibitors. ...
Article
Full-text available
Human epidermal growth factor receptor 2 (HER2)-low breast cancer is defined as breast cancer with an immunohistochemistry (IHC) score of 1+ or 2+ and in situ hybridisation (ISH)-negative. The traditional HER2 classification (negative or positive) has limitations, with only 15%–20% of the breast cancer population being positive and suitable for HER2-targeted therapy. The new clinical study, DESTINY-Breast04, shows that trastuzumab deruxtecan (T-DXd) has a significant effect on advanced HER2-low breast cancers, a classification that accounts for approximately half of the advanced breast cancer population. However, the detection methods and evaluation criteria for HER2-low breast cancer have not yet been standardised, and the toxicity and resistance mechanisms associated with T-DXd therapy are still unclear. This article focuses on these issues and describes the progress and challenges of T-DXd-related therapy in the treatment of advanced breast cancer patients with low HER2 expression.
... Previously, HER2-targeted drugs did not show benefit in this subset of breast cancer as seen in the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-47 phase III trial which showed no benefit for addition of adjuvant trastuzumab to chemotherapy in patients with HER2-negative BC [5]. However, novel anti-HER2 compounds such as trastuzumab deruxtecan (T-Dxd) have shown efficacy across the full spectrum of HER2 expression, including the HER2L subtype, which has expanded treatment options for patients as seen in the randomized phase III clinical trial DESTINY Breast 04 [6]. ...
Article
Full-text available
Background Novel agents have expanded the traditional HER2 definitions to include HER2-Low (HER2L) Breast Cancer (BC). We sought to evaluate the distinct molecular characteristics of HER2L BC to understand potential clinical/biologic factors driving resistance and clinical outcomes. Methods Retrospective analysis was performed on 13,613 BC samples, tested at Caris Life Sciences via NextGen DNA/RNA Sequencing. BC subtypes were defined by IHC/ISH. CODEai database was used to access clinical outcomes from insurance claims data. Results Overall, mutational landscape was similar between HER2L and classical subsets of HR+and HRneg cohorts. TP53 mutations were significantly higher in HRneg/HER2L group vs. HR+/HER2L tumors (p<0.001). A higher mutation rate of PIK3CA was observed in HRneg/HER2L tumors compared to TNBC subtype (p=0.016). PD-L1 positivity was elevated in HRneg/HER2L tumors compared to HR+/HER2L tumors, all p<0.01. Patients with HR+/HER2L tumors treated with CDK4/6 inhibitors had similar OS compared to pts with HR+/HER2-0 (HR=0.89, p=0.012). 27.2% of HR+/HER2L pts had activating PIK3CA mutations. Among HR+PIK3CA mutated tumors, HER2L pts treated with alpelisib showed no difference in OS vs. HER2-0 alpelisib-treated pts (HR=1.23, p=0.517). 13.9% of HER2L TNBC pts were PD-L1⁺. Interestingly, pts with PD-L1⁺ HER2L/HRneg (TNBC) treated with immune checkpoint inhibitors (ICI) showed improved OS than HER2-0 TNBC (HR=0.61, p=0.046). Conclusion Our findings expand the understanding of the molecular profile of the HER2L subgroup and comparison to the classically defined breast cancer subgroups. Genomic risk assessments after progression on novel therapeutics can be assessed to better define implications for mechanisms of resistance.
Article
Background Accurate determination of human epidermal growth factor receptor 2 (HER2) is important for choosing optimal HER2 targeting treatment strategies. HER2‐low is currently considered HER2‐negative, but patients may be eligible to receive new anti‐HER2 drug conjugates. Purpose To use breast MRI BI‐RADS features for classifying three HER2 levels, first to distinguish HER2‐zero from HER2‐low/positive (Task‐1), and then to distinguish HER2‐low from HER2‐positive (Task‐2). Study Type Retrospective. Population 621 invasive ductal cancer, 245 HER2‐zero, 191 HER2‐low, and 185 HER2‐positive. For Task‐1, 488 cases for training and 133 for testing. For Task‐2, 294 cases for training and 82 for testing. Field Strength/Sequence 3.0 T; 3D T1‐weighted DCE, short time inversion recovery T2, and single‐shot EPI DWI. Assessment Pathological information and BI‐RADS features were compared. Random Forest was used to select MRI features, and then four machine learning (ML) algorithms: decision tree (DT), support vector machine (SVM), k ‐nearest neighbors ( k ‐NN), and artificial neural nets (ANN), were applied to build models. Statistical Tests Chi‐square test, one‐way analysis of variance, and Kruskal–Wallis test were performed. The P values <0.05 were considered statistically significant. For ML models, the generated probability was used to construct the ROC curves. Results Peritumoral edema, the presence of multiple lesions and non‐mass enhancement (NME) showed significant differences. For distinguishing HER2‐zero from non‐zero (low + positive), multiple lesions, edema, margin, and tumor size were selected, and the k ‐NN model achieved the highest AUC of 0.86 in the training set and 0.79 in the testing set. For differentiating HER2‐low from HER2‐positive, multiple lesions, edema, and margin were selected, and the DT model achieved the highest AUC of 0.79 in the training set and 0.69 in the testing set. Data Conclusion BI‐RADS features read by radiologists from preoperative MRI can be analyzed using more sophisticated feature selection and ML algorithms to build models for the classification of HER2 status and identify HER2‐low. Level of Evidence 4. Technical Efficacy Stage 2.
Article
Full-text available
The mechanisms of action of and resistance to trastuzumab deruxtecan (T-DXd), an anti-HER2–drug conjugate for breast cancer treatment, remain unclear. The phase 2 DAISY trial evaluated the efficacy of T-DXd in patients with HER2-overexpressing (n = 72, cohort 1), HER2-low (n = 74, cohort 2) and HER2 non-expressing (n = 40, cohort 3) metastatic breast cancer. In the full analysis set population (n = 177), the confirmed objective response rate (primary endpoint) was 70.6% (95% confidence interval (CI) 58.3–81) in cohort 1, 37.5% (95% CI 26.4–49.7) in cohort 2 and 29.7% (95% CI 15.9–47) in cohort 3. The primary endpoint was met in cohorts 1 and 2. Secondary endpoints included safety. No new safety signals were observed. During treatment, HER2-expressing tumors (n = 4) presented strong T-DXd staining. Conversely, HER2 immunohistochemistry 0 samples (n = 3) presented no or very few T-DXd staining (Pearson correlation coefficient r = 0.75, P = 0.053). Among patients with HER2 immunohistochemistry 0 metastatic breast cancer, 5 of 14 (35.7%, 95% CI 12.8–64.9) with ERBB2 expression below the median presented a confirmed objective response as compared to 3 of 10 (30%, 95% CI 6.7–65.2) with ERBB2 expression above the median. Although HER2 expression is a determinant of T-DXd efficacy, our study suggests that additional mechanisms may also be involved. (ClinicalTrials.gov identifier NCT04132960.)
Article
1022 Background: Optimizing sequential use of Antibody Drug Conjugates (ADCs) is an area of unmet need and of rising clinical importance. With the recent approvals of sacituzumab govitecan (SG) for HR+/HER2- and triple negative metastatic breast cancer (MBC) as well as trastuzumab deruxtecan (T-DXd) for HER2-low MBC, many patients are now candidates for multiple ADCs. However, given potential cross-resistance based on antibody target vs payload (Coates et al, Cancer Discov. 2021), optimal sequencing remains uncertain. We evaluated the safety and efficacy of ADC after ADC for patients with HER2 negative MBC. Methods: We included all patients at an academic institution treated with more than one ADC for MBC. Each line of ADC beyond the first was evaluated for presence of the same “antibody target” or “payload” compared to prior ADC. Clinicopathological information was gathered by chart review. We defined “cross-resistance” as progressive disease (PD) at time of first restaging on second ADC. Progression-free survival (PFS) was evaluated as time from start of treatment to disease progression or death from any cause. All PFS estimation was done using the KM method. All pairwise comparisons across ADC were done using a Wilcoxon Rank Sum test to allow for divergences from normality in progression times. Significance was declared as a type I error less than 0.05. Results: A total of 193 patients with MBC were treated with ADCs between August 2014-February 2023. Among these,32 patients were identified as having received more than one ADC (HR+/HER2- = 13, TNBC = 19, HER2 low = 22). Median age at time of second ADC was 57.1 years (range 31.3-88.6) and patients had received a median of 4 lines (range 2-12) of prior treatment before initiation of second ADC. The median PFS on the first ADC used (ADC1) was significantly longer at 7.55 months (95% CI 3.22-10.25) compared to a median of 2.53 months on the second ADC (ADC2) (95% CI 1.38-4.14) (p=0.006). PFS for ADC2 with antibody target change was 3.25 months (95% CI 1.38 months, n/a) compared to 2.30 months with no target change (95% CI 1.38 months, n/a) (p=0.16). At time of first imaging, cross-resistance was present in 17 cases (53.1%), absent in 12 (37.5%), and not evaluable in 3 cases. When the second ADC contained the same antibody target as the first, cross-resistance was present in 9/13 cases (69.2%), compared to 8/16 cases (50.0%) when the second ADC targeted a different tumor antigen. Similarly, differences were noted based on payload switch vs not. Conclusions: This study highlights a subset that had cross-resistance to ADC after ADC, while others had durable responses on latter lines of therapy, particularly if a different antibody target was utilized. Further research is needed to validate these findings and discern mechanisms of clinical resistance to guide optimal sequencing of ADC-based treatment options.
Article
Trastuzumab deruxtecan (T-DXd) is an antibody drug conjugate with a topoisomerase I payload that targets the human epidermal growth factor receptor 2 (HER2). T-DXd is approved for patients with previously treated HER2-positive or HER2-low (immunohistochemistry [IHC] 1+ or IHC 2+/ISH−) metastatic/unresectable breast cancer (BC). In a second-line HER2-positive metastatic BC (mBC) population (DESTINY-Breast03 [ClinicalTrials.gov identifier: NCT03529110 ]), T-DXd demonstrated significantly improved progression-free survival (PFS) over ado-trastuzumab emtansine (12-month rate: 75.8% v 34.1%; hazard ratio, 0.28; P < .001), and in patients with HER2-low mBC treated with one prior line of chemotherapy (DESTINY-Breast04 [ClinicalTrials.gov identifier: NCT03734029 ]), T-DXd demonstrated significantly longer PFS and overall survival than physician's choice chemotherapy (10.1 v 5.4 months; hazard ratio, 0.51; P < .001, and 23.4 v 16.8 months; hazard ratio, 0.64; P < .001, respectively). Interstitial lung disease (ILD) is an umbrella term used for a group of diseases characterized by lung injury including pneumonitis, which can lead to irreversible lung fibrosis. ILD is a well-described adverse event associated with certain anticancer therapies, including T-DXd. An important part of T-DXd therapy for mBC consists of monitoring for and managing ILD. Although information on ILD management strategies is included in the prescribing information, additional information on patient selection, monitoring, and treatment can be beneficial in routine clinical practice. The objective of this review is to describe real-world, multidisciplinary clinical practices and institutional protocols used for patient selection/screening, monitoring, and management related to T-DXd–associated ILD.
Article
Human epidermal growth factor receptor 2-positive breast cancer (HER2+ BC) is an aggressive tumor type, accounting for 15% to 20% of the approximately 300,000 new BC cases in the United States each year. The goal of this review is to discuss the evolving landscape of therapies for HER2+ metastatic BC (mBC). Targeted therapies that have been the standard of care (SOC) for HER2+ mBC for almost a decade have greatly improved patient outcomes. The SOC for the first-line treatment of HER2+ mBC continues to be HER2-targeted monoclonal antibodies (mAbs) + a taxane, but recent updates in the second-line setting favor use of a newer HER2-targeted antibody-drug conjugate (ADC), trastuzumab deruxtecan, versus the prior SOC ADC, trastuzumab emtansine. Numerous options are now available in the third line and beyond, including tyrosine kinase inhibitor (TKI) regimens, newer mAbs, and other ADCs. The optimal course of treatment for individual patients can be guided by location of metastases, prior therapies, concomitant biomarkers, and monitoring and management of adverse events. Ongoing trials will further the evolution of the HER2+ mBC treatment landscape. Furthermore, next-generation ADCs, TKIs, and classes of drugs that have not been approved for the treatment of HER2+ mBC, including immune checkpoint inhibitors and cyclin-dependent kinase 4 and 6 inhibitors, are also being evaluated for their efficacy in the first and second line. Although the influx of new drugs may complicate treatment decisions for physicians, having a multitude of options will undoubtedly further improve patient outcomes and patient-centered care.
Article
Background: In the DESTINY-Breast03 clinical trial, trastuzumab deruxtecan (T-DXd) showed superior progression-free survival and overall survival versus trastuzumab emtansine (T-DM1) and manageable safety in patients with HER2-positive (HER2+) metastatic breast cancer (mBC). Here, patient-reported outcomes (PROs) are reported along with hospitalization data. Patients and methods: Patients in DESTINY-Breast03 were assessed for prespecified PRO measures, including European Organization for Research and Treatment of Cancer quality of life (EORTC-QoL) questionnaires (the oncology-specific EORTC QLQ-C30 and breast cancer-specific EORTC QLQ-BR45) and the generic EuroQol 5-dimension 5-level questionnaire (EQ-5D-5L) visual analogue scale (VAS). Analyses included change from baseline (CFB), time to definitive deterioration (TDD), and hospitalization-related endpoints. Results: EORTC QLQ-C30 baseline global health status (GHS) scores for T-DXd (n = 253) and T-DM1 (n = 260) were similar, with no clinically meaningful change (<10-point CFB) while on either treatment (median treatment duration: T-DXd, 14.3 months; T-DM1, 6.9 months). TDD analyses of QLQ-C30 GHS (primary PRO variable) and all other prespecified PROs (QLQ-C30 subscales, the QLQ-BR45 arm symptoms scale, and the EQ-5D-5L VAS) suggested T-DXd was numerically favored over T-DM1 based on TDD hazard ratios. Of all randomized patients, 18 (6.9%) receiving T-DXd versus 19 (7.2%) receiving T-DM1 were hospitalized, and the median time to first hospitalization was 219.5 versus 60.0 days, respectively. Conclusions: In DESTINY-Breast03, EORTC GHS/QoL was maintained on both therapies throughout treatment, indicating that despite the longer treatment duration with T-DXd versus T-DM1, HRQoL did not worsen on T-DXd. Furthermore, TDD hazard ratios numerically favored T-DXd over T-DM1 in all prespecified variables of interest including pain, suggesting T-DXd may delay time until HRQoL deterioration compared with T-DM1. Median time to first hospitalization was three times longer with T-DXd versus T-DM1. Together with reported improved efficacy and manageable toxicity, these results support the overall benefit of T-DXd for patients with HER2+ mBC.
Article
Background: In the single-arm, phase 2 DESTINY-Breast01 trial, trastuzumab deruxtecan showed robust activity in patients with HER2-positive metastatic breast cancer who were refractory or resistant to trastuzumab emtansine; a population with scarce effective treatments. In DESTINY-Breast02, we aimed to compare the efficacy and safety of trastuzumab deruxtecan with treatment of physician's choice in this patient population. Methods: This randomised, open-label, multicentre, phase 3 trial was conducted at 227 sites (hospitals, university hospitals, clinics, community centres, and private oncology centres) in North America, Europe, Asia, Australia, Brazil, Israel, and Türkiye. Eligible patients were aged 18 years or older, had unresectable or HER2-positive metastatic breast cancer, previously received trastuzumab emtansine, disease progression, an Eastern Cooperative Oncology Group performance status of 0 or 1, and adequate renal and hepatic function. Patients were randomly assigned (2:1) to receive trastuzumab deruxtecan (intravenously at 5·4 mg/kg once every 3 weeks) or treatment of physician's choice using block randomisation. Treatment of physician's choice was either capecitabine (1250 mg/m2; orally twice per day on days 1-14) plus trastuzumab (8 mg/kg intravenously on day 1 then 6 mg/kg once per day) or capecitabine (1000 mg/m2) plus lapatinib (1250 mg orally once per day on days 1-21), with a 21-day schedule. The primary endpoint was progression-free survival based on blinded independent central review in the full analysis set. This study is registered with ClinicalTrials.gov, NCT03523585. Findings: Between Sept 6, 2018, and Dec 31, 2020, 608 patients were randomly assigned to receive trastuzumab deruxtecan (n=406; two did not receive treatment) or treatment of physician's choice (n=202; seven did not receive treatment). 608 (100%) patients were included in the full analysis set. The median age was 54·2 years (IQR 45·5-63·4) in the trastuzumab deruxtecan group and 54·7 years (48·0-63·0) in the treatment of physician's choice group. 384 (63%) patients were White, 603 (99%) were female, and five (<1%) were male. The median follow-up was 21·5 months (IQR 15·2-28·4) in the trastuzumab deruxtecan group and 18·6 months (8·8-26·0) in the treatment of physician's choice group. Median progression-free survival by blinded independent central review was 17·8 months (95% CI 14·3-20·8) in the trastuzumab deruxtecan group versus 6·9 months (5·5-8·4) in the treatment of physician's choice group (HR 0·36 [0·28-0·45]; p<0·0001). The most common treatment-emergent adverse events were nausea (293 [73%] of 404 with trastuzumab deruxtecan vs 73 [37%] of 195 with treatment of physician's choice), vomiting (152 [38%] vs 25 [13%]), alopecia (150 [37%] vs eight [4%]), fatigue (147 [36%] vs 52 [27%]), diarrhoea (109 [27%] vs 105 [54%]), and palmar-plantar erythrodysaesthesia (seven [2%] vs 100 [51%]). Grade 3 or higher treatment-emergent adverse events occurred in 213 (53%) patients receiving trastuzumab deruxtecan versus 86 (44%) receiving treatment of physician's choice; whereas drug-related interstitial lung disease occurred in 42 (10%; including two grade 5 death events) versus one (<1%). Interpretation: DESTINY-Breast02 shows the favourable benefit-risk profile of trastuzumab deruxtecan in patients with HER2 positive metastatic breast cancer, as previously reported in DESTINY-Breast01, and is the first randomised study to show that one antibody-drug conjugate can overcome resistance to a previous one. Funding: Daiichi Sankyo and AstraZeneca.