Abstract
Antibody–drug conjugates (ADCs) are a class of antineoplastic agents whose structure is composed of three main components: a monoclonal antibody (mAB) targeting a specific target antigen, a cytotoxic payload, and a linker binding the antibody to the payload. By combining the specificity of mABs with the high potency of the payloads, ADCs constitute a smart drug delivery system with improved therapeutic index. After recognition and binding of the mAB to its target surface antigen, ADCs are internalized by endocytosis by the tumor cell, releasing the payloads into the cytoplasm, where they exert their cytotoxic activity, eventually leading to cell death. The composition of some of the new ADCs confers additional functional properties that allow expanding their activity to neighboring cells not expressing the target antigen, constituting a valuable strategy to overcome tumor heterogeneity. Some of these ‘off-target effects’, such as the bystander effect, are possibly the mechanism underlying the antitumor activity demonstrated in patients with low expression of the target antigens, which represents an important paradigm shift in anticancer targeted therapy. Three ADCs are currently approved for the treatment of breast cancer (BC); two anti-HER2 (human epidermal growth factor receptor 2) ADCs (trastuzumab emtansine and trastuzumab deruxtecan); and one Trop-2-targeted ADC (sacituzumab govitecan). Based on the unprecedented efficacy data demonstrated by these agents, ADCs have been incorporated as part of standard regimens for all subtypes of advanced BC, as well as for high-risk early HER2-positive BC. Despite the remarkable advances, several hurdles still remain to overcome, including the development of reliable biomarkers for patient selection, prevention, and management of potentially severe toxicities, ADC resistance mechanisms, post-ADC resistance patterns, and optimal treatment sequencing and combinations. In this review, we will summarize the currently available evidence related to the use of these agents, as well as explore the current landscape of ADC development for BC treatment.
Keywords: antibody–drug conjugates, breast cancer, targeted therapy
Background
High rates of adverse events and a narrow therapeutic index 1 were hallmarks of the first chemotherapeutic agents to demonstrate antitumor activity in the early 20th century. Since then, a major drug development ambition in oncology has been the development of compounds that could act on specific tumor targets with minimal or no effect on normal cells, with the promise of improved efficacy and reduced toxicities. 2 This aspiration was largely facilitated by improvements in nuclear acid sequencing technologies that allowed the characterization of malignancies according to molecular alterations, thus examining these aberrations as potential therapeutic targets. Breast cancer (BC) is perhaps one of the best examples of solid malignancies that had their natural history modified by targeted therapies whose development were only made possible by the accumulation of knowledge about tumor biology. 3 However, even in the successful case of anti-HER2 agents for BC therapy, the use of concomitant chemotherapy is often required to optimize antitumor effects. 4 Antibody–drug conjugates (ADC) are a class of anticancer agents developed as an attempt to overcome the major limitations of monoclonal antibodies and cytotoxic compounds through a smart strategy of tumor-specific drug delivery. 5 In this narrative review, we will explore the main characteristics, efficacy data, and safety profile of approved and investigational ADC for the treatment of BC.
Molecular structure and mechanism of action of ADCs
The structure of ADCs is characterized by three core components: a monoclonal antibody (mAB) bound by a molecular linker to a cytotoxic agent (payload). 5 All three elements are important determinants of ADC efficacy and toxicity. Conjugation is the methodology used for the linker fixation on the mAB backbone in which the linker is attached on a stochastic or site-specific manner to one of the mAB’s available amino acids. The average number of drug molecules conjugated per antibody is defined as Drug-Antibody Ratio or ‘DAR’. The conjugation methods used impact the DAR and the homogeneity of the payloads on the mAb, being partially responsible for improvements on ADCs’ therapeutic indexes. 6
The chemical characteristics of the mAB of each ADC are major determinants of its immunogenicity and of the circulating half-life of the drug. 7 The binding of the mAB to its target surface antigen through the fragment antigen-binding (Fab) portion leads to the formation of a mAB-antigen complex, which is then internalized. 8 Internalization of the mAB-antigen complex may occur by several mechanisms, including antigen-dependent caveolin-mediated endocytosis, clathrin-mediated endocytosis, or antigen-independent pinocytosis. 9 In the endosome, acidification or proteolysis triggers the release of the payload in the cell cytoplasm, finally reaching its intracellular target. 5 This mechanism of releasing the payload directly into cells that express the target antigen, in a ‘Trojan horse’ fashion, is responsible for a significant reduction in systemic side effects.
Linkers can be classified as ‘cleavable’ and ‘non-cleavable’, and they are a key component of the structure of any ADC, since they are responsible for reducing potential side effects related to the premature payload release by stabilizing the drug in the circulation. Non-cleavable linkers, such as the thioether linker present in T-DM1, provide a high stability to the compounds and allow a relatively delayed payload release, as this process relies on the lysosomal enzyme degradation of the ADC. 6 Cleavable linkers can be subcategorized as chemically or enzyme-cleavable, and cleavage mechanisms may involve (1) internalization of the ADC-antigen complex by endocytosis, lysosomal processing, and intracellular payload release (‘traditional’ cleavage mechanism) or (2) non-internalizing mechanisms, in which the payload is released extracellularly (e.g. tumor microenvironment or circulation). 10 Non-internalizing mechanisms of payload release are hypothesized to be associated with potentially greater efficacy due to lower dependence on intracellular trafficking process and on high cell-surface antigen expression, but also lower cell selectivity. 10 Thus, a significant part of off-target toxicity of newer generation ADCs has been associated to the premature payload release by cleavable linkers, which are subdivided into four main subclasses (hydrazone, cathepsin B-cleavable, disulfide, and pyrophosphate diester). 6
Payloads are the cytotoxic agents conjugated to the mAB, which are usually highly potent agents associated with excessive toxicity that precludes their use in the unconjugated form. 11 Such a high potency, on the contrary, becomes a major advantage in the ADC context, considering that only a small amount of ADC administrated intravenously will reach tumor cells (as low as 0.001–0.01%) and that the payloads delivered need to be active at nanomolar or even picomolar concentrations.12,13
Although the ‘on-target’ effect (i.e. cytotoxic effect after ADC internalization in cancer cells that express the target antigen) is hypothesized to be the main mechanism of action of ADCs, several features of the components of the ADC structure play a crucial role in other aspects that also contribute to their antitumor activity. 8 In ADCs with cleavable linkers, membrane-permeable hydrophobic payloads may diffuse from the interior of dying cells that endocytosed the ADC to the neighboring cells, on which they can elicit a cytotoxic effect regardless of target antigen expression. 14 This phenomenon known as ‘bystander effect’ is believed to have paramount importance in cases of heterogeneous or low-level expression of the target antigens. In some cases, mABs present in the ADC structure retain Fab-mediated antitumor activity by interfering with pathways initiated by antigens expressed on the cell surface by blocking ligand binding, dimerization, and/or inducing degradation of the target protein.5,14,15 These mechanisms, which are typical of ‘classical’ targeted therapies, may also occur with some ADCs, although they are not their main mechanism of action. Finally, ADCs may elicit immune-mediated antitumor responses by several mechanisms, including (1) antibody-dependent cell-mediated cytotoxicity by the recognition of mAB Fc component by immune cells such as natural killers, (2) antibody-dependent cell-mediated phagocytosis by the interaction with tumor-associated macrophages, and (3) complement-mediated cytotoxicity through C1q binding. 5
ADCs currently approved for BC treatment
Two anti-HER2 ADCs (ado-trastuzumab emtansine and fam-trastuzumab deruxtecan-nxki) and one Trop-2-targeted ADC (sacituzumab govitecan) are currently approved by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of BC (Figure 1, Table 1).
Table 1.
Trial | Study population | Treatment arms | Efficacy results |
---|---|---|---|
Ado-trastuzumab emtansine (T-DM1) | |||
EMILIA
16
(NCT00829166) Phase III |
- HER2-positive aBC - Prior treatment with trastuzumab and a taxane |
T-DM1 versus lapatinib and capecitabine | - mPFS: 9.6 versus 6.4 months (HR 0.65; 95% CI 0.55–0.70) - mOS: 29.9 versus 25.9 months (HR 0.75, 95% CI 0.64–0.88) |
TH3RESA
17
(NCT00829166) Phase III |
- HER2-positive aBC − ⩾2 HER2-directed regimens in the advanced setting |
T-DM1 versus TPC | - mPFS: 6.2 versus 3.3 months (HR 0.52; 95% CI 0.422–0.661) - mOS: 22.7 versus 15.8 months (HR 0.68; 95% CI 0.54–0.85) |
KATHERINE
18
(NCT01772472) Phase III |
- HER2-positive eBC - Residual disease after neoadjuvant therapy - Adjuvant setting |
T-DM1 versus Trastuzumab | - 3 years IDFS: 88.3 versus 77.0% (HR 0.50; 95% CI 0.39–0.64) |
ATEMPT
19
(NCT01853748) Phase II |
- HER2-positive eBC (stage I) - Adjuvant setting |
T-DM1 versus TH | - 5 years IDFS: 97.8% versus 93.4% |
KAITLIN
20
(NCT01966471) Phase III |
- HER2-positive eBC (tumor >2.0 cm or node positive) - Adjuvant setting |
AC/EC followed by: THP versus T-DM1 + P | - 3 years IDFS (ITT population): 94.2% versus 93.1% (HR 0.98; 95% CI, 0.72–1.32) |
KRISTINE
21
(NCT02131064) Phase III |
- HER2-positive eBC (stage II–III) - Neoadjuvant setting |
T-DM1+ P versus TC-HP | - 3 years IDFS: 93% versus 92% (HR 1.11; CI 95%, 0.52–2.40) - 3 years EFS: 85% versus 94% (HR 2.61; CI 95%, 1.36–4.98) |
Trastuzumab deruxtecan (T-DXd) | |||
DESTINY-Breast02
22
(NCT03523585) Phase III |
- HER2-positive aBC - Prior treatment with T-DM1 |
T-DXd versus TPC | - mPFS: 17.8 versus 6.9 months (HR 0.36; 95% CI 0.28–0.45) - mOS: 39.2 versus 26.5 months (HR 0.66; 95% CI 0.50–0.86) |
DESTINY-Breast0323,24 (NCT03529110) Phase III |
- HER2-positive aBC - Prior treatment with trastuzumab and a taxane |
T-DXd versus T-DM1 | - mPFS:28.8 versus 6.8 months (HR 0.33; 95% CI 0.26–0.43) - mOS: NR in either arm (HR 0.64, 95% CI 0.47–0.87) - 24 months OS rate: 77.4% versus 69.9% |
DESTINY-Breast04
25
(NCT03734029) Phase III |
- HER2-low aBC - Prior treatment with 1–2 lines of chemotherapy |
T-DXd versus TPC | - mPFS: 9.9 versus 5.1 months HR 0.50; 95% CI 0.40–0.63) - mOS: 23.4 versus 16.8 months (HR 0.64; 95% CI 0.49–0.84). |
DAISY
26
(NCT04132960) Phase II |
- Cohort 1: HER2-positive aBC - Cohort 2: Her2 low aBC - Cohort 3: HER2 ICH 0 aBC |
T-DXd | - Cohort 1: mPFS 11.1 months (95% CI 8.5–14.4) - Cohort 2: mPFS 6.7 months (95% CI 4.4–8.3) - Cohort 3: mPFS 4.2 months (95% CI 2–5.7) |
TUXEDO-1
27
(NCT04752059) Phase II |
- HER2-positive aBC - Brain metastases newly diagnosed or progressing after prior local treatment |
T-DXd | - ORR: 73.3% (95% CI 48.1–89.1) |
Sacituzumab Govitecan | |||
ASCENT
28
(NCT02574455) Phase III |
- Advanced TNBC - ⩾2 prior lines of chemotherapy |
SG versus TPC | - mPFS: 5.6 versus 1.7 months (HR 0.41; 95% CI, 0.32–0.52) - mOS: 12.1 versus 6.7 months (HR 0.48; 95% CI, 0.38–0.59) |
TROPiCS-0229,30 (NCT03901339) Phase III |
- Hormone receptor positive/HER2-negative aBC - Pretreated with 2–4 prior chemotherapy regimens |
SG versus TPC | - mPFS: 5.5 versus 4.0 months (HR 0.66; 95% CI 0.53–0.83) - mOS: 14.4 versus 11.2 months (HR, 0.79; 95% CI 0.65–0.96) |
aBC, advanced breast cancer; AC/EC, doxorubicin or epirubicin plus cyclophosphamide; ADCs, Antibody–drug conjugates; CI, confidence interval; eBC, early breast cancer; EFS, event-free survival; EMA, European Medicines Agency; HR, hazard ratio; IDFS, invasive disease-free survival; ITT, intention-to-treat; mOS, median overall survival; mPFS, median progression-free survival; NR, not reported; ORR, objective response rate; OS, overall survival; SG, sacituzumab govitecan; TC-HP, docetaxel, carboplatin, and trastuzumab plus pertuzumab; T-DM1 + P, T-DM1 plus pertuzumab; TH, trastuzumab plus paclitaxel; THP, taxane plus trastuzumab plus pertuzumab; TNBC, triple negative breast cancer; TPC, treatment of physician’s choice.
Ado-trastuzumab emtansine
T-DM1 (ado-trastuzumab emtansine) was the first HER2-targeted ADC to be granted approval for the treatment of solid malignancies. This agent combines the anti-HER2 activity of trastuzumab, a humanized anti-HER2 IgG1 mAb, withthe cytotoxic properties of DM1, and a microtubule inhibitor derived from maytansine.Trastuzumab is binded to DM1 through a non-reducible thioether linker(N-succinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate).31,32
In the metastatic setting, the EMILIA trial was the pivotal study demonstrating the efficacy of T-DM1 in the second-line setting. In this study, 991 patients with HER2-positive advanced BC (aBC) previously treated with taxane-based chemotherapy and trastuzumab, were randomized to receive T-DM1 or capecitabine plus lapatinib. T-DM1-based therapy led to significant improvements in all the primary endpoints: median progression-free survival (mPFS) was 9.6 versus 6.4 months (HR 0.65, 95% CI 0.55–0.77, p < 0.001), and in the final descriptive analysis, at a median follow-up of 41.9 months in the control group and 47.8 months in the T-DM1 group, median overall survival (mOS) was 29.9 months versus 25.9 months (HR 0.75, 95% CI 0.64–0.88). 16 These results were confirmed in a more heavily pretreated population by the results of TH3RESA trial, since mPFS (6.2 versus 3.3 months, HR 0.52, 95% CI 0.422–0.661 p = 0.0001) and mOS (22.7 versus 15.8 months, HR 0.68, 95% CI 0.54–0.85, p = 0.0007) were also improved by T-DM1 as compared to the treatment of physician’s choice (TPC) group comprising of chemotherapy (any single agent), hormonal therapy for hormone-receptor-positive disease (single-agent or dual therapy), or HER2-directed therapy (single-agent, dual HER2-targeted therapy, or combination with either single-agent chemotherapy or single-agent hormonal therapy). 17 Hence, until late 2021, T-DM1 was considered the preferred second-line therapy for the treatment of HER2-positive metastatic patients.
In the early setting, the KATHERINE trial led to the approval of T-DM1 as post-neoadjuvant treatment for patients with residual disease after a neoadjuvant treatment. In this phase III trial, 1486 patients who had not achieved a pathological complete response (pCR) after neoadjuvant taxane-based chemotherapy combined with anti-HER2 treatment (with or without anthracyclines) were randomized to receive adjuvant T-DM1 or trastuzumab, both for 14 cycles. At a median follow-up of 41.4 months, the estimated invasive disease-free survival (iDFS) was 88.3% for the T-DM1 arm, compared to 77% for the standard arm (HR 0.50, 95% CI 0.39–0.64, p < 0.001). T-DM1 showed its benefit in every subgroup of patients, notwithstanding pathological nodal status, hormonal expression, age, and type of anti-HER2 therapy used. 18 While the KATHERINE trial has used T-DM1 as an escalation strategy in patients at high risk of relapse, the ATEMPT trial aimed to test if this drug could be used to ‘de-escalate’ adjuvant treatment in patients with stage I HER2-positive disease. In this study, 497 patients were randomized to receive T-DM1 for 17 cycles or paclitaxel and trastuzumab with standard schedule in the adjuvant setting. The objective of the study was to evaluate if T-DM1 was less toxic than standard adjuvant treatment and yielded clinically acceptable iDFS. At a median follow-up of 3.9 years, iDFS were 97.8% and 93.4% for T-DM1 and paclitaxel-trastuzumab, respectively. The incidence of clinically relevant toxicities was similar in both arms (46% versus 47%, p = 0.83) and a higher number of patients (17%) had early T-DM1 discontinuation due to adverse events (AE) as compared to the paclitaxel-trastuzumab arm (6%). 19 The ongoing ATEMPT 2.0 trial is comparing adjuvant paclitaxel and trastuzumab with T-DM1 for six cycles (18 weeks) followed by trastuzumab for 11 cycles (NCT04893109).
T-DM1 activity was also tested in combination with other HER2-targeted agents in the early setting. The phase III KAITLIN trial randomized patients with HER2-positive BC with tumor size >2 cm or node positive to receive adjuvant chemotherapy with anthracyclines followed by pertuzumab and T-DM1 or taxane plus pertuzumab and trastuzumab. No significant difference was seen in terms of iDFS rate at a median follow-up of 57 months, 93.1% (95% CI 91.4–94.7) versus 94.2% (95% CI 92.7–95.8). 20 In the neoadjuvant setting, the phase III KRISTINE trial randomly assigned 444 patients with locally advanced HER2-positive BC to receive neoadjuvant chemotherapy with T-DM1 plus pertuzumab or docetaxel, carboplatin, and trastuzumab plus pertuzumab. Higher pCR rates were seen in patients treated with the standard treatment, as compared to T-DM1 plus pertuzumab (55.7% versus 44.4%; p = 0.016). At a median follow-up of 37 months, the 3-year event-free survival was 85.3% in the T-DM1 and pertuzumab arm, compared to 94.2% in the other arm (HR 2.61, 95% CI 1.36–4.98). No significant differences in iDFS (HR 1.11, 95% CI 0.52–2.40) and OS (HR 1.21, 95% CI 0.37–3.96) were found.18,21
A recent review assessing the combined incidence of AE in patients treated with anti-HER2 agents included data from 20 studies reporting T-DM1- related AE. Among these patients, the most common side effect were decreased platelet count (incidence of 84.6%, with more than 20% of grade ⩾3), hepatotoxicity (53.6%), diarrhea (19.6%), vomiting (19.2%), hypokalemia (10–15%), and cutaneous reactions (7.7%). 33 A pooled analysis of individual data from over 1900 patients treated with T-DM1 in seven clinical trials demonstrated a rate of adverse cardiac events of 3.4%, most of which were characterized by asymptomatic left ventricular ejection fraction (LVEF) drops. 34 Notably, in the TH3RESA trial 7% of patients treated with T-DM1 experienced AE leading to treatment discontinuation, while in the KATHERINE trial, in adjuvant setting, the rate of T-DM1 discontinuation was 28.5%.17,18Figure 2 summarizes the toxicity profile of ADCs currently approved for BC treatment, as reported in the main registration trials.23,28
Trastuzumab Deruxtecan (T-DXd)
Fam-trastuzumab deruxtecan-nxki (T-DXd or DS-8201) is an ADC consisting of humanized anti-HER2 immunoglobulin G1 antibody linked to deruxtecan, a topoisomerase I inhibitor payload derived from exatecan through a tetrapeptide-based cleavable linker. It is characterized by a relevant bystander effect and a high DAR of 8:1 (compared to a DAR of 3.5:1 in T-DM1). 35 In the open-label, single-group DESTINY-Breast01 trial, heavily pretreated patients (median of six previous treatments received) who received T-DXd at the recommended dose (5.4 mg/kg) experienced an ORR of 60.9% (95% CI 53.4–68.0) and a mPFS of 16.4 months (95% CI 12.7 to not reached). 36 The activity of this ADC was further tested in a larger phase III trial (DESTINY-Breast02) comparing T-DXd with TPC in similar later-line patient population (>99% with prior treatment with T-DM1). Primary results of this study confirmed the clinical benefit and superiority of T-DXd over conventional chemotherapy-based regimens in this setting with prolonged PFS (HR, 0.36; 95% CI 0.28–0.45; p < 0.000001) and OS (HR, 0.66; 95% CI 0.50–0.86; p = 0.0021). 37
In the phase III DESTINY-Breast03 trial, 524 patients with HER2-positive aBC pretreated with trastuzumab and a taxane were randomly assigned to receive T-DXd or T-DM1. Patients with newly diagnosed or progressing brain metastases were not eligible. At 12 months, 75.8% of patients treated with T-DXd were alive and without disease progression, compared to 34.1% of those treated with T-DM1 (HR 0.28, 95% CI 0.22–0.37; p < 0.001). Overall response rate (ORR) was 79.7% in the T-DXd arm and 34.2% in the T-DM1 arm. The benefit in terms of PFS was seen in all the subgroups, irrespective of hormone-receptor status, previous treatment with pertuzumab, presence of visceral disease, number of previous lines, or presence of stable brain metastases. 23 Based on the results of this trial, T-DXd has become the preferred second-line regimen for HER2-positive aBC. The updated survival results of this trial were presented in SABCS 2022, confirming the superiority of T-DXd. With a median follow-up of 28.4 months, the risk of death was reduced by 36% (HR, 0.64, 95% CI 0.47–0.87; p = 0.0037) with the median OS not reached in any of the arms. The landmark of 12 months OS rate remained superior in T-DXd (94.1% versus 86.0%) and 24 months OS rate was 77.4% for T-DXd compared to 69.9% for T-DM1. 24
Interestingly, approximately 60% of HER2-negative aBCs express low levels of HER2 (HER2-low), defined as a score of 1+ on immunohistochemistry (IHC) or an IHC score of 2+ and negative results on in situ hybridization (ISH) 38 These tumors were traditionally treated as HER2-negative malignancies due to the inefficacy of previously available HER2-directed therapies to improve their outcomes.39–41 Under the hypothesis that T-DXd could potentially be active in this population particularly through the bystander effect to neighboring tumor cells heterogeneously expressing HER2, 35 T-DXd efficacy was also evaluated in patients with HER2-low aBC in the DESTINY-Breast04 trial. 15 In this phase III trial, patients pretreated for HER2-low aBC were randomized 2:1 to receive T-DXd or TPC (eribulin, capecitabine, gemcitabine, paclitaxel, or nab-paclitaxel). Among 557 randomized patients, median PFS was 9.9 months in the T-DXd arm and 5.1 months in the control arm (HR 0.50, 95% CI 0.40–0.63, p < 0.001), while mOS was increased from 16.8 months to 23.4 months in patients treated with T-DXd (HR 0.64; 95% CI 0.49–0.84, p = 0.001). In the hormone receptor-positive subgroup (88.7%, 494 patients) the mPFS was 10.1 months in the T-DXd arm and 5.4 months in the control arm (HR 0.51, 95% CI, 0.40–0.64, p < 0.001), and mOS was 23.9 versus 17.5 months (HR 0.64, 95% CI, 0.48–0.86, p = 0.003), consistent with the data of overall population. In the hormone-negative subgroup (11.3%, 63 patients), the median mPFS was 8.5 months for patients treated with T-DXd and 2.9 months for patients who have undergone TPC treatment (HR 0.46, 95% CI, 0.24–0.89), mOS was 18.2 and 8.3 months, respectively (HR 0.48, 95% CI 0.24–0.95). 25 However, the number of patients with hormone receptor-negative disease was limited and this data should be seen with caution. Further data of patients with hormone receptor-negative, HER2-low aBC treated with T-DXd were presented in SABCS 2022 with the updated results of the cohort treated with T-DXd in combination with durvalumab from the BEGONIA phase Ib/II trial. In this study, patients treated with the combination in the first-line setting achieved an ORR of 57% and a median PFS of 12.6 months, with a manageable safety profile. 42
The unprecedented results of the DESTINY-Breast04 trial demonstrating the efficacy of T-DXd in patients with HER2-low BC represent a paradigm shift by demonstrating the activity of this agent in a population traditionally considered HER2-negative, which expand its indication for patients with HER2-low tumors traditionally classified as hormone receptor-positive/HER2-negative and triple negative breast cancer (TNBC). Interestingly, new data were presented in SABCS 2022 in the early setting with the presentation of the TRIO-US B-12 TALENT neoadjuvant study. This trial randomized patients with previously untreated, operable invasive early stage, hormone receptor-positive, HER2-low BC measuring >2 cm to receive T-DXd alone (Arm A) or in combination with anastrozole (with gonadotropin-releasing hormone (GnRH) agonist in men and pre/perimenopausal women) (Arm B). The ORR was 68% in arm A and 58% in arm B. At the time of this presentation, some patients were still receiving therapy and data were not fully mature for the primary endpoint (pCR rate at surgery) or for long-term outcomes. 43 In addition to providing efficacy and safety data, window trials such as TRIO-US B-12 TALENT trial provide valuable opportunities for translational research in this setting.
The most common any-grade AEs associated with T-DXd are gastrointestinal (nausea, vomiting, constipation), hematological (anemia, neutropenia, thrombocytopenia), fatigue, and alopecia. 44 Hematological AEs, particularly neutropenia and anemia, and fatigue are also among the most frequent high-grade (grade ⩾3) events and leading causes of dose interruptions due to AEs, which were required in approximately 32% of patients treated with T-DXd. 44 Left ventricular dysfunction was reported in 2.7% (7 of 257) of the patients treated with T-DXd in DESTINY-Breast03; all cases were asymptomatic and low grade (grade 1–2).24,45 Thus, LVEF assessment should be performed before beginning treatment with T-DXd and at regular intervals during treatment. 45 Considering T-DXd’s safety profile, a particular concern relates to interstitial lung disease (ILD) due to reports of high-grade and fatal events. 25 ILD is the most common type of drug-induced lung toxicity and is associated with inflammation and fibrosis of the pulmonary interstitium that may ultimately lead to loss of functional gas exchange. 46 It has been hypothesized that the mechanisms of T-DXd-related ILD may involve target-dependent or target-independent uptake of ADC by normal cells; or direct damage caused by the free payload, either released by premature deconjugation into the circulation or from cells following ADC catabolism.47,48 Preclinical evidence demonstrate that, although T-DXd induces ILD in animal models, those receiving deconjugated DXd did not experience lung toxicity, failing to corroborate the hypothesis of direct lung damage caused by the free payload. 48 Furthermore, the low levels of HER2 expression in alveoli 48 and the occurrence of ILD in patients exposed to non-HER2-targeted ADCs (e.g. HER3-targeted patritumab deruxtecan) 49 undermine the hypothesis that this toxicity is caused by target-dependent ADC uptake. Thus, the accumulated evidence suggests that the development of ILD is predominantly associated with target-independent ADC uptake by normal cells (e.g. immune cells), which is supported by the finding of ADC accumulation in alveolar macrophages (and not in pulmonary epithelial cells) in animal models of T-DXd-induced ILD. 48 In a pooled analysis of nine phase I/II trials with T-DXd including data from 1150 patients (44.3% with BC), the incidence of any-grade ILD was 15.4% (177 patients), of which 77.4% (137/177) were grade 1–2, 8.5% (15/177) were grade 3–4, and 14% (22/177) were grade 5. 50 Importantly, the median time to ILD onset was 5.4 months (range <0.1–46.8 months), with 87% of ILD events occurring within 12 months after starting T-DXd. 50 Table 2 summarizes the incidence and median time to onset of ILD in patients treated with T-DXd in DESTINY-Breast01, DESTINY-Breast02, DESTINY-Breast03, and DESTINY-Breast04 trials. Although ILD most commonly develops in the first 6–12 months of treatment with T-DXd, cases of late onset have been reported up to 2 years from the start of T-DXd, emphasizing the importance of permanent surveillance in patients on treatment with this agent. 25 In this pooled analysis, some characteristics were associated with potentially higher risk of ILD, namely age <65 years, enrollment in Japan, higher doses of T-DXd (>6.4 mg/kg), lower oxygen saturation (<95%), renal impairment (moderate/severe), lung comorbidities, and longer time since initial diagnosis (>4 years). 50 Patients should be closely monitored for signs/symptoms and radiographic changes potentially consistent with this AE as T-DXd administration must be interrupted as soon as a diagnosis of ILD (grade ⩾1) is made. 45 Guidelines recommendations for the management of ILD vary with the AE grade and include dose reductions, administration of corticosteroids, and T-DXd interruption (grade 1/asymptomatic events) or permanent discontinuation in patients who are diagnosed with symptomatic (Grade 2 or greater) ILD/pneumonitis. 45
Table 2.
Trial | Incidence of ILD – n (%) | Median time to ILD onset – days (range) | |||
---|---|---|---|---|---|
Any grade | Grade 1–2 | Grade 3–4 | Grade 5 | ||
DESTINY-Breast01 | 25 (13.6) | 20 (10.9) | 1 (0.5) | 4 (2.2) | 193 (42–535) |
DESTINY-Breast02 | 42 (10.4) | 37 (9.1) | 3 (0.7) | 2 (0.5) | 209 (41–638) |
DESTINY-Breast03 | 27 (10.5) | 25 (9.7) | 2 (0.8) | 0 | 168 (33–507) |
DESTINY-Breast04 | 45 (12.1) | 37 (10) | 5 (1.3) | 3 (0.8) | 129 (26–710) |
ILD, interstitial lung disease.
Sacituzumab govitecan
Sacituzumab govitecan (SG) is an ADC composed of an IgG1 kappa mAb anti-trophoblast cell-surface antigen (Trop-2) bound to SN-38, the active metabolite of irinotecan (topoisomerase I inhibitor), through a hydrolyzable linker.28,51 Trop-2 is a transmembrane calcium signal transducer that is overexpressed in BC, with particularly higher expression in TNBC.52,53 In the phase III ASCENT trial, SG was compared with TPC (eribulin, vinorelbine, capecitabine, or gemcitabine) in patients with metastatic TNBC previously treated with two or more prior standard chemotherapy regimens. 28 When compared with TPC, SG improved mPFS (5.6 versus 1.7 months; HR 0.41, 95% CI 0.32–0.52) among patients without brain metastases, mOS (12.1 versus 6.7 months; HR 0.48, 95% CI 0.38–0.59), and ORR (35% versus 5%). 28
Considering the expression of Trop-2 in other BC subtypes, the activity of SG was evaluated beyond the TNBC population. In the phase III TROPiCS-02 trial, 543 patients with advanced hormone receptor-positive BC pretreated with 2–4 prior systemic therapy regimens (including one prior taxane, one CDK4/6 inhibitor, and one endocrine therapy in any setting) were randomly assigned to SG or TPC (capecitabine, eribulin, vinorelbine, or gemcitabine). Patients with active brain metastases were excluded. 29 SG improved mPFS when compared to TPC (5.5 versus 4.0 months; HR 0.66; 95% CI 0.53–0.83). 29 In the planned second interim analysis, SG also demonstrated a significant improvement in OS (14.4 versus 11.2 months, HR 0.65–0.96). 30 In SABCS 2022 the authors presented an interesting post hoc analysis showing that these improvement were observed regardless of Trop-2 expression. 54
In the ASCENT study, serious treatment-related AE were reported in 15% in the SG arm and in 8% in the TPC arm, although the rate of AE leading to drug discontinuation was similar in both arms. 28 Importantly, SG is associated with a risk of severe neutropenia and diarrhea (and possibly neutropenic colitis). The rates of grade ⩾3 treatment-related AE were the following: neutropenia (51% with SG and 33% with TPC), leukopenia (10% and 5%), diarrhea (10% and <1%), anemia (8% and 5%), and febrile neutropenia (6% and 2%). 28 In the TROPiCS-02 trial, the safety profile of SG was consistent with previous trials with the drug. 30
Incorporation of ADCs into BC clinical practice guidelines
Different ADCs have been endorsed as standard therapy for the treatment of all BC subtypes by regulatory authorities around the world. However, the positioning of these agents in treatment algorithms remains an important challenge, especially considering differences in the registration trials and the absence of data on the use of new ADCs after prior ADC exposure. Based on the available data, we propose a simplified algorithm for incorporating these agents into the management of metastatic breast cancer (Figure 3).
For HER2-positive BC, T-DXd has become the preferred second-line therapy based on DESTINY-Breast03 data.27 T-DXd is currently approved by the FDA and EMA for patients with advanced HER2-positive BC who have received one or more prior anti-HER2-based regimens.44,56 Based on data from the TH3RESA study, T-DM1 remains a valid FDA- and EMA-approved later-line treatment option, although there are no data demonstrating its efficacy after prior T-DXd treatment. 17
For TNBC, SG is a standard clinical option for patients with pretreated disease based on the results of the ASCENT trial. 28 SG is approved by the FDA and the EMA for the treatment of patients with advanced TNBC who have received two or more prior systemic therapies, at least one of them for metastatic disease.57,58 Patients with TNBC and HER2-low disease who have been previously treated with chemotherapy for metastatic disease (or have had disease recurrence during or within 6 months after completing adjuvant chemotherapy) might also be considered for T-DXd based on DESTINY-Breast04 data.36 T-DXd is approved by the FDA and the EMA for the treatment of patients with HER2-low (TNBC or hormone receptor-positive) advanced BC who have received prior chemotherapy in the metastatic setting or developed disease recurrence during or within 6 months of completing adjuvant chemotherapy.44,59
Finally, patients with hormone receptor-positive disease who have previously received 2–4 lines of systemic therapy (including one prior taxane, one CDK4/6 inhibitor, and one endocrine therapy in any setting) should be considered for SG based on the results of the TROPiCS-02 trial. 30 SG is approved by the FDA for the treatment of patients with advanced hormone receptor-positive (HER2-zero or HER2-low) BC who have received endocrine-based therapy and at least two additional systemic therapies in the metastatic setting. 60 T-DXd might also be considered as a treatment option for patients with hormone receptor-positive and HER2-low BC, who have previously received at least one line of endocrine therapy and one line of chemotherapy based on the results of DESTINY-Breast04. 25 There are no head-to-head comparative data in these two agents in patients with HER2-low disease. Therefore, toxicity profile and patients’ preferences should be considered in the decision-making process.
For patients with brain metastases (BM), the subgroup analysis of patients with stable BM at baseline in DESTINY-Breast03, confirms benefit in PFS of T-DXd (HR 0.38, 95% CI 0.23–0.64), 23 position this drug as a second-line therapies option after THP also in case of stable central nervous system (CNS) involvement. 27 Of particular interest, the activity of T-DXd in patients with active BM was not evaluated in DESTINY-Breast03 trial because only patients with stable BM were included in the study. 23 The TUXEDO trial was a phase II, single-arm study assessing the activity of T-DXd in patients with HER2-positive aBC after exposure to trastuzumab and pertuzumab and with BM that were previously untreated or progressing after previous local therapy. 27 In this trial, the intracranial ORR was 73.3% (95% CI 48.1–89.1). 27 Although limited by the small sample size (15 patients) and absence of comparator arm, these data demonstrate the CNS activity of T-DXd and suggest a potential role of T-DXd also in patients with active central nervous system metastases.
HER2-targeted ADCs in clinical development for the treatment of BC
Building on the success achieved with T-DM1 and T-DXd, HER2 is currently the most intensively studied ADC target in BC, setting an extremely dynamic drug development environment. In this highly competitive field, several strategies are being explored, including innovations in the structure and components of ADCs, often with the aim of achieving tumor responses not only in HER2-positive tumors, but also in the highly prevalent HER2-low population, in which T-DXd has demonstrated unprecedented benefits. Table 3 summarizes the characteristics, efficacy, and safety profile of HER2-targeted ADCs in clinical development for the treatment of BC (non-exhaustive list).
Table 3.
ADC | Structure | Efficacy | Most frequent adverse events | Development status |
---|---|---|---|---|
Trastuzumab duocarmazine (SYD985)61,62 | - mAB: trastuzumab - Linker: cleavable - Payload: duocarmycin/seco-DUBA (alkylating) - DAR: 2.8:1 |
Phase III (TULIP trial): - mPFS: 7.0 versus 4.9 months in the SYD985 and TPC arms, respectively (HR 0.64, 95% CI 0.49–0.84) - mOS: 20.4 versus 16.3 months (HR 0.83, 95% CI 0.62–1.09) |
- Most common AE were eye toxicity and fatigue. - ILD reported in 7.6% (22 patients), with two grade 5 events. |
License application is under review by regulatory authorities. |
A16663,64 | - mAB: trastuzumab variant - Linker: cleavable (valine-citrulline) - Payload: duostatin-5 (auristatin) - DAR: 2:1 |
Phase I: - ORR: 73.9%, mPFS 12.3 months in 4.8 mg/kg cohort - ORR: 68.6%, mPFS 9.4 months in 6.0 mg/kg cohort |
- Any-grade AE: corneal epitheliopathy, blurred - Vision, peripheral sensory neuropathy - AE grade ⩾3: corneal epitheliopathy, blurred vision, ulcerative keratitis |
A phase II trial (NCT05346328) in patients with HER2-positive aBC is ongoing. |
Disitamab vedotin (RC48)65,66 | - mAB: hertuzumab - Linker: cleavable (valine-citruline) - Payload: MMAE (auristatin) - DAR: 4:1 |
Pooled analysis of C001 and C003 CANCER phase I trials: HER2-positive cohort: - ORR 22.2, 42.9, and 40.0%, for 1.5, 2.0, and 2.5 mg/kg dose levels, respectively. - mPFS 4.0, 5.7, and 6.3 months, respectively. - HER2-low cohort (2.0 mg/kg): - ORR: 39.6% - mPFS: 5.7 months |
- Any-grade AE: AST/ALT increase, hypoesthesia, neutropenia - AE grade ⩾3: neutropenia, GGT increased, fatigue |
Ongoing studies in HER2-positive (NCT03500380) and HER2-low aBC (NCT04400695). |
ARX78867–69 | - mAB: anti-HER2 mAB - Linker: non-cleavable - Payload: amberstatin (AS269) - DAR: 1.9:1 |
ACE-Breast-01 - ORR: 74% (1.5 mg/kg) - ACE-Pan tumor-01 - ORR: 67% (1.5 mg/kg) |
- Any-grade AE: fatigue, neutropenia, thrombocytopenia - AE grade ⩾3: ocular toxicity, pneumonitis, fatigue |
Ongoing trials in HER2-positive aBC (ACE-Breast-03, NCT04829604), HER2-low aBC (NCT05018676), patients with brain metastases (NCT05018702). ARX788 development is currently paused due to strategic decision. |
PF-06804103 70 | - mAB: trastuzumab variant - Linker: cleavable - Payload: Aur0101 (auristatin) - DAR: 4:1 |
Phase I: - ORR 52.4% (dose ⩾3 mg/kg) |
- Any-grade AE: alopecia, fatigue, and neuropathy - AE grade ⩾3: arthralgia, neuropathy, and myalgia |
Discontinued. |
MEDI4276 71 | - mAB: bispecific ADC targeting subdomains 2 and 4 of the HER2 receptor - Linker: cleavable - Payload: tubulysin (AZ13599185) - DAR: 4:1 |
Phase I: - ORR: 9.4% |
- Any-grade AE: nausea, fatigue, and AST/ALT increase - AE grade ⩾3: AST/ALT increased, diarrhea |
Discontinued. |
XMT-152272,73 | - mAB: HT-19 - Linker: cleavable (cysteine) - Payload: AF-HPA (auristatin) - DAR: 12:1 |
Phase I: - DCR 83% - ORR: 16.7% |
- Any-grade AE: increased liver enzymes, fatigue, and nausea | Discontinued. |
ALT-P7 (HM2/MMAE) 74 | - mAB: trastuzumab variant - Linker: cleavable (cysteine) - Payload: MMAE (auristatin) - DAR: 2:1 |
Phase I: - DCR: 77.3% - ORR: 13.3% |
- Any-grade AE: myalgia, fatigue, and sensory neuropathy - AE grade ⩾3: neutropenia |
No ongoing trials. |
Zanidatamab zovodotin (ZW49) 75 | - mAB: bispecific ADC targeting subdomains 2 and 4 of HER2 - Linker: cleavable (valine-citruline) - Payload: zovodotin (auristatin) - DAR: NR |
Phase I: - DCR: 50% - ORR: 13% |
- Any-grade AE: keratitis, alopecia, and diarrhea - AE grade ⩾3: infusion-related reaction, neutropenia |
Enrollment in this study still ongoing (NCT03821233). |
MRG002 76 | - mAB: trastuzumab variant - Linker: cleavable (cysteine) - Payload: MMAE (auristatin) - DAR: 3.8:1 |
Phase I (HER2-low): - DCR: 75.5% - ORR: 34.7% |
- Any-grade AE: neutropenia, leukopenia, AST/ALT increase - AE grade ⩾3: neutropenia |
Ongoing enrollment in Phase I trial (NCT03821233). Ongoing trials in HER2-positive aBC (NCT04924699, NCT05263869) |
FS-1502 77 | - mAB: trastuzumab - Linker: cleavable - Payload: MMAF (auristatin) - DAR: 2:1 |
Phase Ia: - ORR: 66.7% (doses ⩾1.0 mg/kg) |
- Any-grade AE: NR - AE grade ⩾3: hypokalemia, thrombocytopenia |
Ongoing phase III trial (NCT05755048) |
aBC, advanced breast cancer; ADC, antibody–drug conjugate; AE, adverse event; ALT, alanine aminotransferase; AST: aspartate aminotransferase; CI, confidence interval; DAR, drug-to-antibody ratio; DCR, disease control rate; F; HR, hazard ratio; ILD, interstitial lung disease; mAB, monoclonal antibody; mPFS, median progression-free survival; mOS, median overall survival; NR, not reported; MMAE, Monomethyl auristatin E; MMAF, Monomethyl auristatin; ORR, objective response rate.
Trastuzumab duocarmazine is a novel HER2-targeted ADC that, although not yet approved, is in a more advanced stage of development. This agent, also known as SYD985 is an anti-HER2 ADC consisting of trastuzumab, a cleavable linker, and a duocarmycin (vc-seco-DUBA) payload. 61 In the phase I trial, 39 patients with different tumor types (including 26 with BC, among which 17 were HER2-positive) with at least 1 + HER2 expression assessed by immunohistochemistry were enrolled in the dose-escalation phase and 146 patients (50 with HER2-positive and 49 with HER2-low BC) were enrolled in the dose-expansion phase. ORR was 33% (95% CI 20.4–48.4) in patients with HER2-positive BC, 28% (13.8–46.8) in HER2-low, hormone-receptor positive, and 40% (16.3–67.6) in HER2-low hormone-receptor negative BC. 61 In the phase III TULIP study, 437 patients with advanced HER2-positive disease, pretreated with two or more lines of therapy were randomly assigned in a 2:1 ratio to receive SYD985 or TPC. 62 Median PFS was 7.0 months in the SYD985 arm and 4.9 months in the TPC arm (HR 0.64, 95% CI 0.49–0.84, p = 0.002). At the first interim analysis, no significant difference was seen between the two arms in terms of mOS (20.4 versus 16.3 months, HR 0.83, 95% CI 0.62–1.09, p = 0.153). 62 The most common AE of any-grade reported in the TULIP trial were conjunctivitis (38.2%), keratitis (38.2%), and fatigue (33.3%), with 52.8% of patients treated with SYD985, experiencing grade ⩾3 AE. 62 Notably, to mitigate the eye toxicity risk, patients with prior keratitis were excluded, lubricating eye drops were administered prophylactically, eye examinations were regularly performed by an ophthalmologist. Nevertheless, eye toxicity remains the most common AE in this trial, reported by 78.1% of patients with 21.2% of grade ⩾3, requiring dose reductions in 22.9% and drug discontinuation in 20.8% of cases. The incidence of ILD was 7.6% (22 patients), with 2.4% (7 patients) being of grade ⩾3 and 0.7% (2 patients) of grade 5. This AE led to treatment discontinuation in 5.2% of cases, despite the exclusion of patients with history of pneumonitis and the implementation of a full diagnostic workup for new or worsening respiratory symptoms. 62 SYD985 license application is currently under review by regulatory authorities.
In terms of their structure (Table 3), most anti-HER2 ADCs in earlier stages of development for BC treatment are composed of: (1) a HER2-targeted mAB, which can be either trastuzumab (or a variation) or a novel mAB targeting one or more HER2 subdomains (e.g. bispecific mABs targeting subdomains 2 and 4 of HER2); (2) a cleavable linker (with the exception of ARX788), 67 (3) a payload more commonly belonging to the class of microtubule inhibitors, with DAR ranging from 2 to 12 molecules of payload per mAB. Among the innovations explored in these new compounds we can mention the wide incorporation of cleavable linkers and innovative bispecific mABs targeting two epitopes in subdomains 2 and 4 of the HER2 receptor, which results in crosslinking followed by internalization of the complex and release of the payload after the cleavage of the linker. 75
Although cross-trial comparisons are not possible considering the heterogeneity of study designs and the different populations included in each study, ORR ranging from 13% to as high as 75% were observed with these agents. In terms of their safety profiles, hematological AEs (including low- and high-grade events) are among the most common incident toxicities observed with several of these agents, which is probably related to the payloads’ mechanisms of action.65,68,76 Importantly, the safety profiles of each of these agents should be closely monitored as some patients might experience high grade (or fatal AEs) and also because the toxicity of these agents is not easily predictable based on the ADC’s structure. For instance, some important toxicities of the new ADCs were not commonly observed either with T-DM1 or other HER2-targeted agents, namely ocular toxicities, an important class of AEs observed with trastuzumab duocarmazine, A166, ARX788, and other agents.62,63,68
Beyond HER2: investigational ADCs exploring other targets for the treatment of BC
Although considerable research efforts have been employed in the search for new therapeutic targets in the last decades, it was only with the approval of SG for the treatment of TNBC in 2021 that an ADC targeting a non-HER2 target was incorporated into clinical practice. 28 The benefit demonstrated with this anti-Trop-2 agent proved the concepts that a new generation of ADC could effectively target proteins that are highly expressed in tumors, even if they are not part of hyperactivated oncogenic driver pathways. These findings further boosted the development of ADC targeting new targets, many of which with promising initial results in all BC subtypes. Table 4 summarizes data from ADCs exploring other (non-HER2) targets for the treatment of BC, several of which use antimicrotubule payloads and cleavable linkers with variable DAR.
Table 4.
ADC | Structure | Efficacy | Most frequent adverse events | Development status |
---|---|---|---|---|
Patritumab deruxtecan (HER3-DXd, U3-1402)49,78 |
- Target: HER3 - mAB: patritumab - Linker: cleavable (tetrapeptide based) - Payload: deruxtecan (camptothecin) - DAR: 8:1 |
aBC: Phase I/II: - ORR: 42.9% (HER2-positive) - ORR: 30.1% (HR-positive/HER2-negative) - ORR: 22.6% (TNBC) - eBC: - ORR: 45% (HR-positive/HER2-negative) |
- Any-grade AE: nausea, neutropenia, and thrombocytopenia - AE grade ⩾3: neutropenia, thrombocytopenia, and anemia. There was one grade 5 ILD event. |
Ongoing phase II trials: - aBC (NCT04699630) - aBC HER3-positive, HR-positive (ICARUS-BREAST, NCT04965766) - eBC HR-positive/HER2-negative (VALENTINE, NCT05569811) |
Datopotamab deruxtecan (Dato-DXd, DS-1062a)79,80 |
Target: Trop-2 mAB: datopotamab Linker: cleavable (tetrapeptide based) Payload: deruxtecan (camptothecin) DAR: 4:1 |
Phase I (TROPION-PanTumor01) TNBC: - Overall population: ○ ORR: 32% ○ mPFS: 4.3 months (95% CI: 3.0–7.3) ○ mOS: 12.9 months (95% CI: 10.1–14.7) - Patients with no prior exposure to Topo 1 inhibitor-based ADC: ○ ORR: 44% ○ mPFS: 7.3 months (95% CI: 3.0-NE) ○ mOS: 14.3 months (95% CI: 10.5-NE) HR-positive/HER2-negative: - ORR: 29% |
- Any-grade AE: nausea, stomatitis, alopecia, and fatigue - AE grade ⩾3: stomatitis, fatigue, mucosal inflammation, and eye toxicity |
Ongoing phase III trials: - TROPION-Breast01 (NCT05104866): advanced TNBC, second/third lines - TROPION-Breast02 (NCT05374512): advanced TNBC first line - TROPION-Breast03 (NCT05629585): early TNBC with RD after neoadjuvant therapy |
Ladiratuzumab vedotin (SGN-LIV1A) 81 | - Target: LIV-1 - mAB: hLIV22 - Linker: cleavable (dipeptide) - Payload: MMAE (auristatin) - DAR: 4:1 |
Phase I (SGNLVA-001) - ORR: 28% (1.25 mg/kg) |
- Any-grade AE: fatigue, nausea, and peripheral neuropathy - AE grade ⩾3: neutropenia, fatigue, and hyperglycemia |
Ongoing studies: - neoadjuvant (NCT01042379) - aBC, in combination with pembrolizumab (SGNLVA-002, NCT03310957) 82 - aBC, in combination with atezolizumab (NCT03424005) 83 |
Cofetuzumab pelidotin (PF-06647020) 84 |
- Target: PTK7 - mAB: hu6M024 - Linker: cleavable (valine-citruline) - Payload: auristatin-0101 - DAR: 4:1 |
Phase I: - ORR: 21% (TNBC) |
- Any-grade AE: nausea, alopecia, and fatigue - AE grade ⩾3: neutropenia |
No ongoing trials in breast cancer. |
Anti-CA6-DM4 immunoconjugate (SAR566658)85,86 | - Target: CA6 - mAB: DS6 - Linker: cleavable (disulfide) - Payload: DM4 (maytansine) - DAR: 1:1 |
Phase I
85
: - ORR: 60% (190 and 90 mg/m2 on day 1 and day 8) - ORR: 35% (150 and 120 mg/m2 q2 w.) Phase II: terminated due to safety reasons. 86 |
- Any-grade AE: keratopathy, fatigue, and peripheral neuropathy - AE grade ⩾3: NR |
No ongoing trials in breast cancer. |
Praluzatamab ravtansine (CX-2009)87,88 | - Target: CD166 - mAB: CD166-targed mAB - Linker: cleavable - Payload: DM4 (maytansine) - DAR: 3.5:1 |
Phase I/II
87
: - ORR: 9% (HR-positive) Phase II 88 : - Arm A (HR-positive/HER2-negative): ORR: 14.9%, mPFS: 11.4 weeks - Arm B (TNBC): ORR: <10% |
- Any-grade AE: blurred vision, nausea, and fatigue - AE grade ⩾3: ocular and neuropathic events |
No ongoing trials in breast cancer. |
aBC, advanced breast cancer; ADC, antibody–drug conjugate; AE, adverse event; CI, 95% confidence interval; DAR, drug-to-antibody ratio; eBC, early breast cancer; HR-positive, hormone receptor-positive; ILD, interstitial lung disease; mAB, monoclonal antibody; mPFS, median progression-free survival; mOS: median overall survival; NR, not reported; ORR, objective response rate; q2w, every 2 weeks; RD, residual disease; TNBC, triple negative breast cancer.
Datopotamab deruxtecan (Dato-DXd, DS-1062a) is another Trop-2 targeted ADC composed of a human IgG1 mAb that is enzymatically conjugated by a cleavable tetrapeptide-based linker with a topoisomerase I inhibitor (DXd) payload. 89 Relying on the high expression of Trop-2 in epithelial carcinomas and in the high potency of the payload (DXd), the activity of Dato-DXd was evaluated in patients with BC. In the BC cohort of the phase I TROPION-PanTumor01 study, including 43 patients with pretreated advanced TNBC the ORR was 39% (and 52% among those not previously treated with SG, another topoisomerase 1 inhibitor-based ADC). 90 The rates of AE of any grade and grade ⩾3 were 95% and 35%, respectively. The most commonly reported AE were nausea (58%), stomatitis (53%), alopecia (35%), vomiting (35%), and fatigue (33%). 90 The ongoing phase III TROPION-Breast01 (NCT05104866) and TROPION-Breast02 (NCT05374512) will assess the activity of this agent in patients with pretreated and treatment-naïve metastatic TNBC, respectively.
In addition to HER2, other members of the human epidermal growth factor (EGFR/HER) family have been explored as novel ADC targets. HER3 is a membrane receptor with little intrinsic tyrosine kinase activity, that commonly participates in the activation of oncogenic signaling via PI3K/Akt pathway and Src kinase by the formation of heterodimers with other receptor tyrosine kinases, including HER2.91,92 Patritumab deruxtecan (HER3-DXd, U3-1402) is a HER3-directed ADC comprising a fully human anti-HER3 immunoglobulin G1 (IgG1) mAb linked to a topoisomerase I inhibitor payload (deruxtecan) via a tetrapeptide-based cleavable linker. 91 U31402-A-J101 was a phase I/II, first-in-human study of HER3-DXd in patients with HER3-expressing aBC. The ORR in the HER2-positive (n = 14) and hormone receptor-positive (n = 113) disease and TNBC (n = 53) were 42.9%, 30.1%, and 22.6%, respectively. 49 The rate of grade ⩾3 AEs was 71.4%, with the most common being neutropenia (39.6%), thrombocytopenia (30.8%), anemia (18.7%), and leukopenia (18.1%). Importantly, the rate of treatment-related ILD was 6.6%, including one grade 5 event. 49 The activity of HER3-DXd was also evaluated in the early setting. SOLTI TOT-HER3 study enrolled patients with untreated hormone receptor-positive/HER2-negative operable (⩾1 cm) BC (divided in four cohorts with different levels of ERBB3 mRNA expression) to receive a single dose of HER3-DXd. The ORR was 45% among the 77 evaluable patients. 78 Ongoing studies (NCT04965766, NCT04699630) will provide additional data about the efficacy and safety of this agent.
Interestingly, updated results from the phase Ib/II BEGONIA trial presented at SABCS 2022 showed that patients with advanced TNBC treated with the combination of Dato-DXd with durvalumab (anti-PD-L1) as first-line therapy had an ORR of 79% (95% CI: 61–91) with 2/33 patients (6%) having a complete response and 24/33 (73%), a partial response. 93 Although preliminary, these data suggest the potential benefit of combining ADCs with immune checkpoint inhibitors, a strategy also being tested with other agents. Ladiratuzumab vedotin (SGN-LIV1A) is an LIV-1 directed ADC composed of mAb targeting LIV-1, a protease-cleavable dipeptide linker, and a microtubule polymerization inhibitor payload (MMAE). 94 LIV-1 is a transmembrane protein with metalloproteinase activity and downstream target of STAT3 that is expressed in a moderate/high level in the majority of BCs.95,96 Ongoing studies are investigating the combination of this agent with other pembrolizumab (NCT03310957) and atezolizumab (NCT03424005).
Predictive biomarkers for ADCs
With an increasing number of agents demonstrating clinical efficacy, predictive biomarkers will be an essential part of the incorporation of these new agents into clinical practice. As of now, HER2 expression is the only biomarker available for patient selection, although the demonstration of the activity of some ADCs in patients with low HER2 expression represented a complete paradigm shift which will require further and in-depth exploration.
T-DM1 antitumor activity is closely related with HER2 expression. The efficacy of this agent was evaluated in patients who did not fulfill traditional HER2 positivity criteria (IHC +3 or ISH positive) in a centralized reanalysis of HER2 status in patients enrolled in a phase II trial with T-DM1. In this population, the activity associated with T-DM1 was significantly limited (ORR of 4.8% and median PFS of 2.6 months). 41 Evidence suggesting a correlation between the levels (and homogeneity) of HER2 expression and the response to T-DM1 is also available in the early setting. In the neoadjuvant KRISTINE study, locoregional progression was more frequent in the T-DM1 than in the trastuzumab, pertuzumab, and chemotherapy arm (7% versus <1%, respectively), and the majority (66%) of patients progressing under T-DM1 were IHC + 2 with ISH amplified. 97 Additionally, in a neoadjuvant study including patients with HER2-positive disease receiving T-DM1 plus pertuzumab, no pathologically complete response occurred in patients with heterogeneous HER2 expression. Notably, 75% of these cases were IHC2+ with ISH amplified. 98
Although T-DXd improved clinical outcomes of patients with HER2-positive and HER2-low disease, the activity of this agent also correlates with HER2 expression levels.23,25,26 The relationship between different levels of HER2 expression and the efficacy of T-DXd was prospectively explored in DAISY trial. In this phase II study, patients with aBC pretreated with at least one chemotherapy regimen in the advanced setting were divided in three cohorts: (1) IHC 3+ or IHC 2+ with ISH amplified, (2) IHC 2 + and ISH not amplified or IHC1+, and (3) IHC 0. The ORR to T-DXd according to HER2 expression were 71, 37.5, and 30%, respectively (p < 0.0001) and the median PFS were 11.1, 6.7, and 4.2 months, respectively (p < 0.0001), suggesting a correlation between HER2 expression and response to this agent. 26 Moreover, nonresponse to T-DXd was correlated with a high percentage of HER2-zero cells and their spatial distribution (wide areas of HER2-zero cells distant to HER2 3+ cells). 26 In this study, no driver alterations in baseline biopsies were associated with primary resistance to T-DXd. A higher frequency of mutations in the DNA repair gene SLX4 were identified in biopsies performed at resistance (20%, 4/20 patients) than at baseline (2%, 2/88 patients), suggesting that this gene may be implicated in secondary resistance to T-DXd. Interestingly, 13 out of 20 (65%, 95% CI: 40.8–84.6) patients had a decrease in the baseline HER2 expression at the time of progression on T-DXd. Similarly, trastuzumab duocarmazine not only showed activity in HER2-positive BC, but also in HER2-low disease, with an ORR of 32% and a median PFS of 4 months in the metastatic setting. 61 Among 48 patients with HER2-low BC treated with RC48, ORR was 40% and median PFS was 5.7 months. 65 Based on these results, an ongoing phase III study is currently testing RC48 in patients with metastatic HER2-low BC (NCT04400695). The ongoing DESTINY-Breast06 (NCT04494425) will assess the efficacy of T-DXd compared with TPC in patients with HER2-low aBC, including those with ultralow expression (IHC 0+). A particularly important challenge in this setting is the limitation of current immunohistochemistry assays to differentiate HER2-low from HER2-zero, as they were not designed for this purpose. 99
Considering the variety of agents with activity demonstrated for the treatment of HER2-positive aBC,100,101 the identification of predictive biomarkers will be a key factor to guide optimal treatment sequencing. Importantly, under the pressure of HER2-targeted agents, HER2 expression levels may vary during the disease course, with some primary HER2-positive tumors relapsing as HER2-negative or HER2-low, and conversely.102,103 Thus, it can be hypothesized that the levels of HER2 expression and its distribution among different metastatic foci may be significantly modified by the prior treatment with HER2-targeted ADCs, which could possibly impact the efficacy of subsequent therapies. Molecular imaging has been successfully used to explore intra-/interpatient heterogeneity in HER2 mapping of metastatic disease and to identify patients unlikely to benefit from HER2-targeted agents. In the ZEPHIR trial, the patterns of HER2 radio-labeled tracer uptake assessed by HER2-positron emission tomography (PET-CT) with 82 Zr-trastuzumab have been shown to be a promising strategy for understanding tumor heterogeneity in aBC and for selecting patients who will/will not benefit from T-DM1-based therapy. 104
Beyond the levels of HER2 expression, the presence of HER2-mutations appears to be a relevant predictor of ADC efficacy. Interestingly, preclinical data suggest that, regardless of the presence of HER2 overexpression, ubiquitination, and internalization of the receptor are the main mechanisms driving endocytosis and subsequent antitumor activity of anti-HER2 ADCs. 105 Thus, HER2 mutations that increase the rate of receptor internalization and trafficking may induce responses upon exposure to ADCs, regardless of their intrinsic dependence on HER2 signaling for cell growth and/or survival. 105 These findings are supported by clinical data demonstrating high ORR from T-DM1 and T-DXd in HER2-mutated metastatic NSCLC (ORR 44% and 55%, respectively).106,107 Interestingly, in hormone receptor-positive disease (but not in TNBC), ERBB2 and luminal-related genes are more expressed in HER2-low than HER2 IHC 0. 38 Ongoing studies are assessing the efficacy of T-DXd in patients with HER2-mutated solid tumors, including BC (NCT04639219).
Finally, the accumulated evidence suggest that the benefit derived from SG is observed regardless of the levels of Trop-2 expression in hormone receptor-positive and TNBC and the assessment of this biomarker is currently not recommended for patient selection.54,108 Thus, the clinically significant activity of T-DXd and SG in patients with low expressions of HER2 and Trop-2 demonstrate that IHC should not be the only biomarker for treatment selection.
Conclusions and future perspectives
BC is a markedly heterogeneous disease, a characteristic intrinsically related to resistance to cytotoxic agents and traditional targeted therapy. ADCs are a class of potent anticancer agents characterized by a smart form of targeted drug delivery that have demonstrated unprecedented efficacy in the treatment of early and advanced BC and have challenged paradigms by demonstrating efficacy in populations with low expression of target antigens. 25
Despite all the progress achieved, several challenges still remain to be overcome. There is still a long way to improve efficacy, better prevention, and management AEs, and increase understanding of biomarkers and resistance mechanisms (payload- vs target-driven). Some of these hurdles may be overcome by ongoing studies exploring (1) new therapeutic targets (e.g. EGFR, RON, EpCAM); (2) bispecific ADCs, inhibiting multiple pathways or modulating the interface between tumor and immune cells; (3) dual-payload ADCs; and (4) potential synergistic interactions between ADCs and other agents such as immune checkpoint inhibitors.82,83,109–112 If future research in this field continues properly addressing these important questions, ADCs could potentially replace cytotoxic and traditional targeted agents as the preferred therapeutic strategy for treatment of BC.
Acknowledgments
Not applicable.
Footnotes
ORCID iDs: Guilherme Nader-Marta https://orcid.org/0000-0001-7864-3637
Véronique Debien https://orcid.org/0000-0003-1165-925X
Ahmad Awada https://orcid.org/0000-0001-7412-9163
Contributor Information
Guilherme Nader-Marta, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Rue Meylemeersch, 90, Anderlecht, Brussels 1070, Belgium.
Chiara Molinelli, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium.
Véronique Debien, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium.
Diogo Martins-Branco, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium.
Philippe Aftimos, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium.
Evandro de Azambuja, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium.
Ahmad Awada, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium.
Declarations
Ethics approval and consent to participate: Not applicable.
Consent for publication: Not applicable.
Author contributions: Guilherme Nader-Marta: Conceptualization; Methodology; Project administration; Writing – original draft.
Chiara Molinelli: Conceptualization; Methodology; Writing – original draft.
Véronique Debien: Conceptualization; Methodology; Visualization; Writing – original draft.
Diogo Martins-Branco: Conceptualization; Methodology; Writing – review & editing.
Philippe Aftimos: Methodology; Resources; Writing – review & editing.
Evandro De Azambuja: Methodology; Resources; Supervision; Writing – review & editing.
Ahmad Awada: Conceptualization; Methodology; Project administration; Resources; Supervision; Writing – review & editing.
Funding: The authors received no financial support for the research, authorship, and/or publication of this article.
Competing interests: GNM: Travel grants from Roche, AstraZeneca and Bayer. CM: Honoraria from Novartis and Lilly. VD: None. DMB: Honoraria from Daiichi Sankyo, Novartis, Merck Sharp & Dohme, Janssen, Pfizer, Angelini, and AstraZeneca; meeting/travel grants from Novartis, Merck Sharp & Dohme, LEO Farmacêuticos, Ipsen, Janssen, Roche, Laboratórios Vitória, and Gilead Sciences; institutional grants from Novartis and F. Hoffmann-La Roche Ltd. PA: Consulting: Boehringer Ingelheim, Macrogenics, Roche, Novartis, Amcure, Servier, G1 Therapeutics, Radius, Deloitte; Honoraria: Synthon, Amgen, Novartis, Gilead; Travel grants: Amgen, MSD, Pfizer, Roche; and Research funding to my institution: Roche. EdA: reports speaking fee from Lilly; support to attend medical conferences from Lilly, Novartis, Roche, Genetic, and Istituto Gentili. AA: reports support from Roche, Lilly, Amgen, EISAI, BMS, Pfizer, Novartis, MSD, Genomic Health, Ipsen, AstraZeneca, Bayer, LEO Pharma, Merck, Daiichi, Seattle Genetics, and Pierre Fabre.
Availability of data and materials: Not applicable.
References
- 1.Alnaim L. Therapeutic drug monitoring of cancer chemotherapy. J Oncol Pharm Pract 2007; 13: 207–221. [DOI] [PubMed] [Google Scholar]
- 2.Gerber DE. Targeted therapies: a new generation of cancer treatments. Am Fam Physician GP 2008; 77: 311–319. [PubMed] [Google Scholar]
- 3.Wang J, Xu B. Targeted therapeutic options and future perspectives for HER2-positive breast cancer. Signal Transduct Target Ther 2019; 4: 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Inoue K, Nakagami K, Mizutani M, et al. Randomized phase III trial of trastuzumab monotherapy followed by trastuzumab plus docetaxel versus trastuzumab plus docetaxel as first-line therapy in patients with HER2-positive metastatic breast cancer: the JO17360 Trial Group. Breast Cancer Res Treat 2010; 119: 127–136. [DOI] [PubMed] [Google Scholar]
- 5.Drago JZ, Modi S, Chandarlapaty S. Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat Rev Clin Oncol 2021; 18: 327–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Tsuchikama K, An Z. Antibody-drug conjugates: recent advances in conjugation and linker chemistries. Protein Cell 2018; 9: 33–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Fu Z, Li S, Han S, et al. Antibody drug conjugate: the ‘biological missile’ for targeted cancer therapy. Signal Transduct Target Ther 2022; 7: 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Khongorzul P, Ling CJ, Khan FU, et al. Antibody-drug conjugates: a comprehensive review. Mol Cancer Res 2020; 18: 3–19. [DOI] [PubMed] [Google Scholar]
- 9.Kovtun YV, Goldmacher VS. Cell killing by antibody-drug conjugates. Cancer Lett 2007; 255: 232–240. [DOI] [PubMed] [Google Scholar]
- 10.Bargh JD, Isidro-Llobet A, Parker JS, et al. Cleavable linkers in antibody-drug conjugates. Chem Soc Rev 2019; 48: 4361–4374. [DOI] [PubMed] [Google Scholar]
- 11.Garrison MA, Hammond LA, Geyer CE, Jr, et al. A Phase I and pharmocokinetic study of exatecan mesylate administered as a protracted 21-day infusion in patients with advanced solid malignancies. Clin Cancer Res 2003; 9: 2527–2537. [PubMed] [Google Scholar]
- 12.Beck A, Goetsch L, Dumontet C, et al. Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov 2017; 16: 315–337. [DOI] [PubMed] [Google Scholar]
- 13.Epenetos AA, Snook D, Durbin H, et al. Limitations of radiolabeled monoclonal antibodies for localization of human neoplasms. Cancer Res 1986; 46: 3183–3191. [PubMed] [Google Scholar]
- 14.Ogitani Y, Hagihara K, Oitate M, et al. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci 2016; 107: 1039–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Redman JM, Hill EM, AlDeghaither D, et al. Mechanisms of action of therapeutic antibodies for cancer. Mol Immunol 2015; 67: 28–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Diéras V, Miles D, Verma S, et al. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol 2017; 18: 732–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Krop IE, Kim S-B, González-Martín A, et al. Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomised, open-label, phase 3 trial. Lancet Oncol 2014; 15: 689–699. [DOI] [PubMed] [Google Scholar]
- 18.von Minckwitz G, Huang C-S, Mano MS, et al. Trastuzumab emtansine for residual invasive HER2-Positive breast cancer. New Engl J Med 2019; 380: 617–628. [DOI] [PubMed] [Google Scholar]
- 19.Tolaney SM, Tayob N, Dang C, et al. Adjuvant trastuzumab emtansine versus paclitaxel in combination with trastuzumab for Stage I HER2-Positive Breast Cancer (ATEMPT): a randomized clinical trial. J Clin Oncol 2021; 39: 2375–2385. [DOI] [PubMed] [Google Scholar]
- 20.Krop IE, Im SA, Barrios C, et al. Trastuzumab emtansine plus pertuzumab versus taxane plus trastuzumab plus pertuzumab after anthracycline for high-risk human epidermal growth factor receptor 2-positive early breast cancer: the Phase III KAITLIN Study. J Clin Oncol 2022; 40: 438–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hurvitz SA, Martin M, Symmans WF, et al. Neoadjuvant trastuzumab, pertuzumab, and chemotherapy versus trastuzumab emtansine plus pertuzumab in patients with HER2-positive breast cancer (KRISTINE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol 2018; 19: 115–126. [DOI] [PubMed] [Google Scholar]
- 22.Krop I, Park YH, Kim S-B, et al. Abstract GS2-01: GS2-01 trastuzumab deruxtecan vs physician’s choice in patients with HER2+ unresectable and/or metastatic breast cancer previously treated with trastuzumab emtansine: primary results of the randomized, phase 3 study DESTINY-Breast02. Cancer Res 2023; 83: GS2–01. [Google Scholar]
- 23.Cortes J, Kim S, Chung W. Trastuzumab deruxtecan (T-DXd) vs trastuzumab emtansine (T-DM1) in patients (Pts) with HER2+ metastatic breast cancer (mBC): results of the randomized phase III DESTINY-Breast03 study. Ann Oncol 2021; S1283–S1346. [Google Scholar]
- 24.Hurvitz SA, Hegg R, Chung W-P, et al. Trastuzumab deruxtecan versus trastuzumab emtansine in patients with HER2-positive metastatic breast cancer: updated results from DESTINY-Breast03, a randomised, open-label, phase 3 trial. J Lancet 2023; 401: 105–117. [DOI] [PubMed] [Google Scholar]
- 25.Modi S, Jacot W, Yamashita T. Trastuzumab deruxtecan in previously treated HER2-Low advanced breast cancer. New Engl J Med 2022; 387: 9–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mosele MF, Lusque A, Dieras V, et al. LBA1 unraveling the mechanism of action and resistance to trastuzumab deruxtecan (T-DXd): biomarker analyses from patients from DAISY trial. Ann Oncol 2022; 33: S123–S147. [Google Scholar]
- 27.Bartsch R, Berghoff AS, Furtner J, et al. Trastuzumab deruxtecan in HER2-positive breast cancer with brain metastases: a single-arm, phase 2 trial. Nat Med 2022; 28: 1840–1847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bardia A, Hurvitz SA, Tolaney SM, et al. Sacituzumab Govitecan in metastatic triple-negative breast cancer. New Engl J Med 2021; 384: 1529–1541. [DOI] [PubMed] [Google Scholar]
- 29.Rugo HS, Bardia A, Marmé F, et al. Sacituzumab Govitecan in hormone receptor–Positive/human epidermal growth factor receptor 2–Negative metastatic breast cancer. J Clin Oncol 2022; 40: 3365–3376. [DOI] [PubMed] [Google Scholar]
- 30.Rugo HS, Bardia A, Marmé F, et al. LBA76 overall survival (OS) results from the phase III TROPiCS-02 study of sacituzumab govitecan (SG) vs treatment of physician’s choice (TPC) in patients (pts) with HR+/HER2- metastatic breast cancer (mBC). Ann Oncol 2022; 33: S1386–S869. [Google Scholar]
- 31.Lewis Phillips GD, Li G, Dugger DL, et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res 2008; 68: 9280–9290. [DOI] [PubMed] [Google Scholar]
- 32.Junttila TT, Li G, Parsons K, et al. Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancer. Breast Cancer Res Treat 2011; 128: 347–356. [DOI] [PubMed] [Google Scholar]
- 33.Perez EA, Dang C, Lee C, et al. Incidence of adverse events with therapies targeting HER2-positive metastatic breast cancer: a literature review. Breast Cancer Res Treat 2022; 194: 1–11. [DOI] [PubMed] [Google Scholar]
- 34.Pondé N, Ameye L, Lambertini M, et al. Trastuzumab emtansine (T-DM1)-associated cardiotoxicity: pooled analysis in advanced HER2-positive breast cancer. Eur J Cancer 2020; 126: 65–73. [DOI] [PubMed] [Google Scholar]
- 35.Ogitani Y, Aida T, Hagihara K, 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 Rev 2016; 22: 5097–5108. [DOI] [PubMed] [Google Scholar]
- 36.Modi S, Saura C, Yamashita T, et al. Trastuzumab deruxtecan in previously treated HER2-Positive breast cancer. New Engl J Med 2020; 382: 610–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.André F, Hee Park Y, Kim S-B, et al. Trastuzumab deruxtecan versus treatment of physician’s choice in patients with HER2-positive metastatic breast cancer (DESTINY-Breast02): a randomised, open-label, multicentre, phase 3 trial. J Lancet 2023; 401: 1773–1785. [DOI] [PubMed] [Google Scholar]
- 38.Schettini F, Chic N, Brasó-Maristany F, et al. Clinical, pathological, and PAM50 gene expression features of HER2-low breast cancer. NPJ Breast Cancer 2021; 7: 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fehrenbacher L, Cecchini RS, Geyer CE, Jr, et al. NSABP B-47/NRG Oncology Phase III randomized trial comparing adjuvant chemotherapy with or without trastuzumab in high-risk invasive breast cancer negative for HER2 by FISH and with IHC 1+ or 2+. J Clin Oncol 2020; 38: 444–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gianni L, Lladó A, Bianchi G, et al. Open-label, phase II, multicenter, randomized study of the efficacy and safety of two dose levels of pertuzumab, a human epidermal growth factor receptor 2 dimerization inhibitor, in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol 2010; 28: 1131–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Burris Ha, 3rd, Rugo HS, Vukelja SJ, et al. Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy. J Clin Oncol 2011; 29: 398–405. [DOI] [PubMed] [Google Scholar]
- 42.Schmid P, Wysocki P, Park YH, et al. Abstract PD11-08: PD11-08 trastuzumab deruxtecan (T-DXd) + durvalumab (D) as first-line (1L) treatment for unresectable locally advanced/metastatic hormone receptor-negative (HR−), HER2-low breast cancer: updated results from BEGONIA, a phase 1b/2 study. Cancer Res 2023; 83: PD11-08. [Google Scholar]
- 43.Bardia A, Hurvitz S, Press MF, et al. Abstract GS2-03: GS2-03 TRIO-US B-12 TALENT: neoadjuvant trastuzumab deruxtecan with or without anastrozole for HER2-low, HR+ early stage breast cancer. Cancer Res 2023; 83: GS2–03. [Google Scholar]
- 44.Enhertu EMA. Summary of Product Characteristics. European Medicines Agency, https://www.ema.europa.eu/en/medicines/human/EPAR/enhertu (2020, accessed 28 March 2023).
- 45.Rugo HS, Bianchini G, Cortes J, et al. Optimizing treatment management of trastuzumab deruxtecan in clinical practice of breast cancer. ESMO Open 2022; 7: 100553. DOI: 10.1016/j.esmoop.2022.100553 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tarantino P, Modi S, Tolaney SM, et al. Interstitial lung disease induced by Anti-ERBB2 antibody-drug conjugates: a review. JAMA Oncol 2021; 7: 1873–1881. [DOI] [PubMed] [Google Scholar]
- 47.de Goeij BE, Lambert JM. New developments for antibody-drug conjugate-based therapeutic approaches. Curr Opin Immunol 2016; 40: 14–23. [DOI] [PubMed] [Google Scholar]
- 48.Kumagai K, Aida T, Tsuchiya Y, et al. Interstitial pneumonitis related to trastuzumab deruxtecan, a human epidermal growth factor receptor 2-targeting Ab-drug conjugate, in monkeys. Cancer Sci 2020; 111: 4636–4645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Krop IE, Masuda N, Mukohara T, et al. Results from the phase 1/2 study of patritumab deruxtecan, a HER3-directed antibody-drug conjugate (ADC), in patients with HER3-expressing metastatic breast cancer (mBC). J Clin Oncol 2022; 40: 1002–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Powell CA, Modi S, Iwata H, et al. Pooled analysis of drug-related interstitial lung disease and/or pneumonitis in nine trastuzumab deruxtecan monotherapy studies. ESMO Open 2022; 7: 100554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Goldenberg DM, Cardillo TM, Govindan SV, et al. Trop-2 is a novel target for solid cancer therapy with sacituzumab govitecan (IMMU-132), an antibody-drug conjugate (ADC). Oncotarget 2015; 6: 22496–22512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zaman S, Jadid H, Denson AC, et al. Targeting Trop-2 in solid tumors: future prospects. Onco Targets Ther 2019; 12: 1781–1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Aslan M, Hsu E-C, Garcia-Marques FJ, et al. Oncogene-mediated metabolic gene signature predicts breast cancer outcome. NPJ Breast Cancer 2021; 7: 141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Rugo H, Bardia A, Marmé F, et al. Abstract GS1-11: Sacituzumab Govitecan (SG) vs treatment of Physician’s Choice (TPC): efficacy by trop-2 expression in the TROPiCS-02 study of patients (Pts) with hr+/her2– Metastatic Breast Cancer (mBC). Cancer Res 2023; 83: GS1–11. [Google Scholar]
- 55.Lu YS, Mahidin EIBM, Azim H, et al. Abstract GS1-10: primary results from the randomized Phase II RIGHT choice trial of premenopausal patients with aggressive HR+/HER2− advanced breast cancer treated with ribociclib + endocrine therapy vs physician’s choice combination chemotherapy. Cancer Res 2023; 83: GS1–10. [Google Scholar]
- 56.Center for Drug Evaluation and Research. FDA grants regular approval to fam-trastuzumab deruxtecan-nxki for breast cancer. FDA, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-regular-approval-fam-trastuzumab-deruxtecan-nxki-breast-cancer (2022, accessed 22 May 2023).
- 57.Center for Drug Evaluation and Research. FDA grants regular approval to sacituzumab govitecan for triple-negative breast cancer. FDA, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-regular-approval-sacituzumab-govitecan-triple-negative-breast-cancer (2021, accessed 22 May 2023).
- 58.Trodelvy EMA. European Medicines Agency, https://www.ema.europa.eu/en/medicines/human/EPAR/trodelvy (2021, accessed 22 May 2023).
- 59.Center for Drug Evaluation and Research. FDA approves fam-trastuzumab deruxtecan-nxki for HER2-low breast cancer, FDA (2022, accessed 22 May 2023). [Google Scholar]
- 60.Center for Drug Evaluation and Research. FDA approves sacituzumab govitecan-hziy for HR-positive breast cancer. FDA, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-sacituzumab-govitecan-hziy-hr-positive-breast-cancer (2023, accessed 22 May 2023).
- 61.Banerji U, van Herpen CML, Saura C, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol 2019; 20: 1124–1135. [DOI] [PubMed] [Google Scholar]
- 62.Manich CS, O’Shaughnessy J, Aftimos PG, et al. Primary outcome of the phase III SYD985.002/TULIP trial comparing vic- trastuzumab duocarmazine to physician’s choice treatment in patients with pre-treated HER2-positive locally advanced or metastatic breast cancer. Ann Oncol Epub ahead of print 2021. DOI: 10.1016/j.annonc.2021.08.2088. [DOI] [Google Scholar]
- 63.Hu X, Zhang J, Liu R, et al. Updated results and biomarker analyses from the phase I trial of A166 in patients with HER2-expressing locally advanced or metastatic solid tumors. J Clin Oncol 2022; 40: 1037–1037. [Google Scholar]
- 64.Liu Y, Lian W, Zhao X, et al. A first in-human study of A166 in patients with locally advanced/metastatic solid tumors which are HER2-positive or HER2-amplified who did not respond or stopped responding to approved therapies. J Clin Oncol 2020; 38: 1049–1049. [Google Scholar]
- 65.Wang J, Liu Y, Zhang Q, et al. RC48-ADC, a HER2-targeting antibody-drug conjugate, in patients with HER2-positive and HER2-low expressing advanced or metastatic breast cancer: a pooled analysis of two studies. J Clin Oncol 2021; 39: 1022–1022. [Google Scholar]
- 66.Yao X, Jiang J, Wang X, et al. A novel humanized anti-HER2 antibody conjugated with MMAE exerts potent anti-tumor activity. Breast Cancer Res Treat 2015; 153: 123–133. [DOI] [PubMed] [Google Scholar]
- 67.Barok M, Le Joncour V, Martins A, et al. ARX788, a novel anti-HER2 antibody-drug conjugate, shows anti-tumor effects in preclinical models of trastuzumab emtansine-resistant HER2-positive breast cancer and gastric cancer. Cancer Lett 2020; 473: 156–163. [DOI] [PubMed] [Google Scholar]
- 68.Hurvitz SA, Park H, Frentzas S, et al. Safety and unique pharmacokinetic profile of ARX788, a site-specific ADC, in heavily pretreated patients with HER2-overexpresing solid tumors: results from two phase 1 clinical trials. J Clin Oncol 2021; 39: 1038–1038. [Google Scholar]
- 69.Abdollahpour-Alitappeh M, Lotfinia M, Gharibi T, et al. Antibody-drug conjugates (ADCs) for cancer therapy: strategies, challenges, and successes. J Cell Physiol 2019; 234: 5628–5642. [DOI] [PubMed] [Google Scholar]
- 70.Meric-Bernstam F, Calvo E, Moreno V, et al. A phase I dose escalation study evaluating the safety and tolerability of a novel anti-HER2 antibody-drug conjugate (PF-06804103) in patients with HER2-positive solid tumors. J Clin Oncol 2020; 38: 1039–1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Pegram MD, Hamilton EP, Tan AR, et al. First-in-Human, phase 1 dose-escalation study of biparatopic Anti-HER2 antibody-drug conjugate MEDI4276 in patients with HER2-positive advanced breast or gastric cancer. Mol Cancer Ther 2021; 20: 1442–1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hamilton EP, Barve MA, Bardia A, et al. Phase 1 dose escalation of XMT-1522, a novel HER2-targeting antibody-drug conjugate (ADC), in patients (pts) with HER2-expressing breast, lung and gastric tumors. J Clin Oncol 2018; 36: 2546–2546. [Google Scholar]
- 73.Le Joncour V, Martins A, Puhka M, et al. A novel Anti-HER2 antibody-drug conjugate XMT-1522 for HER2-Positive breast and gastric cancers resistant to trastuzumab emtansine. Mol Cancer Ther 2019; 18: 1721–1730. [DOI] [PubMed] [Google Scholar]
- 74.Park YH, Ahn HK, Kim J-Y, et al. First-in-human phase I study of ALT-P7, a HER2-targeting antibody-drug conjugate in patients with HER2-positive advanced breast cancer. J Clin Oncol 2020; 38: 3551–3551. [Google Scholar]
- 75.Jhaveri K, Han H, Dotan E, et al. 460MO Preliminary results from a phase I study using the bispecific, human epidermal growth factor 2 (HER2)-targeting antibody-drug conjugate (ADC) zanidatamab zovodotin (ZW49) in solid cancers. Ann Oncol 2022; 33: S749–S750. [Google Scholar]
- 76.Jiang Z, Sun T, Wang X, et al. A multiple center, open-label, single-arm, phase II clinical trial of MRG002, an HER2-targeted antibody-drug conjugate, in patients with HER2-low expressing advanced or metastatic breast cancer. J Clin Oncol 2022; 40: 1102–1102.35015587 [Google Scholar]
- 77.Li Q, Wang X, Cheng Y, et al. Abstract P4-01-07: FS-1502, an anti-HER2 ADC, in patients with HER2-Expressing advanced solid tumors: a Phase 1a dose-escalation study. Cancer Res 2023; 83: P4-01-07. [Google Scholar]
- 78.Prat A, Falato C, Pare Brunet L, et al. LBA3 patritumab deruxtecan (HER3-DXd) in early-stage HR+/HER2- breast cancer: final results of the SOLTI TOT-HER3 window of opportunity trial. Ann Oncol 2022; 33: S164–S174. [Google Scholar]
- 79.Meric-Bernstam F, Krop I, Juric D, et al. Abstract PD13-08: PD13-08 Phase 1 TROPION-PanTumor01 study evaluating datopotamab deruxtecan (Dato-DXd) in unresectable or metastatic hormone Receptor–Positive/HER2–Negative breast cancer (BC). Cancer Res 2023; 83: PD13-08. [Google Scholar]
- 80.Bardia A, Krop I, Meric-Bernstam F, et al. Abstract P6-10-03: datopotamab deruxtecan (Dato-DXd) in Advanced Triple-Negative Breast Cancer (TNBC): updated results from the Phase 1 TROPION-PanTumor01 Study. Cancer Res 2023; 83: 6–10. [Google Scholar]
- 81.Tsai M, Han HS, Montero AJ, et al. 259P weekly ladiratuzumab vedotin monotherapy for metastatic triple-negative breast cancer. Ann Oncol 2021; 32: S474–S515. [Google Scholar]
- 82.Han HS, Alemany CA, Brown-Glaberman UA, et al. SGNLVA-002: single-arm, open label phase Ib/II study of ladiratuzumab vedotin (LV) in combination with pembrolizumab for first-line treatment of patients with unresectable locally advanced or metastatic triple-negative breast cancer. J Clin Oncol 2019; 37: TPS1110–TPS1110. [Google Scholar]
- 83.Yardley DA, Abu-Khalaf M, Boni V, et al. Abstract OT2-06-04: MORPHEUS: a phase Ib/II trial platform evaluating the safety and efficacy of multiple cancer immunotherapy combinations in patients with hormone receptor–positive and triple-negative breast cancer. Cancer Res 2019; 79: OT2-06-04. [Google Scholar]
- 84.Maitland ML, Sachdev JC, Sharma MR, et al. First-in-Human study of PF-06647020 (Cofetuzumab Pelidotin), an antibody-drug conjugate targeting protein tyrosine kinase 7, in advanced solid tumors. Clin Cancer Rev 2021; 27: 4511–4520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Gomez-Roca CA, Boni V, Moreno V, et al. A phase I study of SAR566658, an anti CA6-antibody drug conjugate (ADC), in patients (Pts) with CA6-positive advanced solid tumors (STs)(NCT01156870). J Clin Oncol 2016; 34: 2511–2511. [Google Scholar]
- 86.EudraCT Number 2016-001962-27 – Clinical trial results – EU Clinical Trials Register. 2019, https://www.clinicaltrialsregister.eu/ctr-search/trial/2016-001962-27/results (accessed 12 October 2022).
- 87.Boni V, Fidler MJ, Arkenau H-T, et al. Praluzatamab Ravtansine, a CD166-Targeting antibody-drug conjugate, in patients with advanced solid tumors: an open-label Phase I/II Trial. Clin Cancer Rev 2022; 28: 2020–2029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Miller K, Tolaney S, Emens LA, et al. Abstract P4-01-15: preliminary results from a phase 2 study of praluzatamab ravtansine (CX-2009) in patients with advanced breast cancer (aBC). Cancer Res 2023; 83: P4-01-15. [Google Scholar]
- 89.Okajima D, Yasuda S, Maejima T, et al. Datopotamab deruxtecan, a novel TROP2-directed antibody-drug conjugate, demonstrates potent antitumor activity by efficient drug delivery to tumor cells. Mol Cancer Ther 2021; 20: 2329–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Krop I, Juric D, Shimizu T, et al. Abstract GS1-05: Datopotamab deruxtecan in advanced/metastatic her2- breast cancer: results from the phase 1 TROPION-PanTumor01 study. Cancer Res 2022; 82: GS1–05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Koyama K, Ishikawa H, Abe M, et al. Patritumab deruxtecan (HER3-DXd), a novel HER3 directed antibody drug conjugate, exhibits in vitro activity against breast cancer cells expressing HER3 mutations with and without HER2 overexpression. PLoS One 2022; 17: e0267027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lyu H, Han A, Polsdofer E, et al. Understanding the biology of HER3 receptor as a therapeutic target in human cancer. Acta Pharm Sin B 2018; 8: 503–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Schmid P, Wysocki P, Ma C, et al. Abstract PD11-09: PD11-09 Datopotamab deruxtecan (Dato-DXd) + durvalumab (D) as first-line (1L) treatment for unresectable locally advanced/metastatic triple-negative breast cancer (a/mTNBC): updated results from BEGONIA, a phase 1b/2 study. Cancer Res 2023; 83: PD11-09. [Google Scholar]
- 94.Nagayama A, Vidula N, Ellisen L, et al. Novel antibody-drug conjugates for triple negative breast cancer. Ther Adv Med Oncol 2020; 12: 1758835920915980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Taylor KM, Hiscox S, Nicholson RI. Zinc transporter LIV-1: a link between cellular development and cancer progression. Trends Endocrinol Metab 2004; 15: 461–463. [DOI] [PubMed] [Google Scholar]
- 96.Sussman D, Smith LM, Anderson ME, et al. SGN-LIV1A: a novel antibody-drug conjugate targeting LIV-1 for the treatment of metastatic breast cancer. Mol Cancer Ther 2014; 13: 2991–3000. [DOI] [PubMed] [Google Scholar]
- 97.Hurvitz SA, Martin M, Jung KH, et al. Neoadjuvant trastuzumab emtansine and pertuzumab in human epidermal growth factor receptor 2-Positive breast cancer: three-year outcomes from the Phase III KRISTINE Study. J Clin Oncol 2019; 37: 2206–2216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Filho OM, Viale G, Stein S, et al. Impact of HER2 heterogeneity on treatment response of early-stage HER2-positive breast cancer: phase II neoadjuvant clinical trial of T-DM1 combined with pertuzumab. Cancer Discov 2021; 11: 2474–2487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Fernandez AI, Liu M, Bellizzi A, et al. Examination of low ERBB2 protein expression in breast cancer tissue. JAMA Oncol 2022; 8: 1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gennari A, André F, Barrios CH, et al. ESMO clinical practice guideline for the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann Oncol 2021; 32: 1475–1495. [DOI] [PubMed] [Google Scholar]
- 101.Nader-Marta G, Martins-Branco D, de Azambuja E. How we treat patients with metastatic HER2-positive breast cancer. ESMO Open 2022; 7: 100343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Tarantino P, Gandini S, Nicolò E, et al. Evolution of low HER2 expression between early and advanced-stage breast cancer. Eur J Cancer 2022; 163: 35–43. [DOI] [PubMed] [Google Scholar]
- 103.Miglietta F, Griguolo G, Bottosso M, et al. Author Correction: evolution of HER2-low expression from primary to recurrent breast cancer. NPJ Breast Cancer 2021; 7: 149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gebhart G, Lamberts LE, Wimana Z, et al. Molecular imaging as a tool to investigate heterogeneity of advanced HER2-positive breast cancer and to predict patient outcome under trastuzumab emtansine (T-DM1): the ZEPHIR trial. Ann Oncol 2016; 27: 619–624. [DOI] [PubMed] [Google Scholar]
- 105.Li BT, Michelini F, Misale S, et al. HER2-Mediated internalization of Cytotoxic agents inERBB2Amplified or mutant lung cancers. Cancer Discov 2020; 10: 674–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Li BT, Shen R, Buonocore D. Ado-Trastuzumab emtansine for patients with HER2-Mutant lung cancers: results from a phase II Basket Trial. J Clin Oncol 2018; 36: 2532–2537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Li BT, Smit EF, Goto Y, et al. Trastuzumab Deruxtecan in HER2-Mutant Non-Small-Cell Lung Cancer. N Engl J Med 2022; 386: 241–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bardia A, Tolaney SM, Punie K, et al. Biomarker analyses in the phase III ASCENT study of sacituzumab govitecan versus chemotherapy in patients with metastatic triple-negative breast cancer. Ann Oncol 2021; 32: 1148–1156. [DOI] [PubMed] [Google Scholar]
- 109.Yamazaki CM, Yamaguchi A, Anami Y, et al. Antibody-drug conjugates with dual payloads for combating breast tumor heterogeneity and drug resistance. Nat Commun 2021; 12: 3528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zhang C, Sheng W, Al-Rawe M, et al. EpCAM- and EGFR-Specific antibody drug conjugates for triple-negative breast cancer treatment. Int J Mol Sci 2022; 23: 6122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Yao H-P, Suthe SR, Hudson R, et al. Antibody-drug conjugates targeting RON receptor tyrosine kinase as a novel strategy for treatment of triple-negative breast cancer. Drug Discov Today 2020; 25: 1160–1173. [DOI] [PubMed] [Google Scholar]
- 112.Torres ETR, Emens LA. Emerging combination immunotherapy strategies for breast cancer: dual immune checkpoint modulation, antibody-drug conjugates and bispecific antibodies. Breast Cancer Res Treat 2022; 191: 291–302. [DOI] [PubMed] [Google Scholar]