Next Article in Journal
The Clinical Outcomes and Safety of Sacituzumab Govitecan in Heavily Pretreated Metastatic Triple-Negative and HR+/HER2− Breast Cancer: A Multicenter Observational Study from Turkey
Previous Article in Journal
Correction: Będkowska et al. What We Know and Do Not Yet Know About the Canine Model of Lymphoma in Human Medicine—The Current State of Knowledge. Cancers 2025, 17, 596
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Role of ADCs in Brain Metastases and Primary Brain Tumors: Insight and Future Directions

by
Francesco Bruzzone
1,2,†,
Chiara Barigazzi
1,2,†,
Antonio Di Muzio
1,2,
Isabel Tallarico
1,2,
Angelo Dipasquale
1,
Agnese Losurdo
1,2,
Pasquale Persico
1,2,
Pierina Navarria
3,
Federico Pessina
2,4,
Armando Santoro
1,2,‡ and
Matteo Simonelli
1,2,*,‡
1
Department of Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
2
Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
3
Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
4
Department of Neurosurgery, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors also contributed equally to this work.
Cancers 2025, 17(9), 1591; https://doi.org/10.3390/cancers17091591
Submission received: 18 March 2025 / Revised: 28 April 2025 / Accepted: 2 May 2025 / Published: 7 May 2025
(This article belongs to the Section Cancer Metastasis)

Simple Summary

Primary and secondary brain tumors are historically treated with loco-regional approaches such as surgery or radiation therapy. Recently, new pharmacological agents like tyrosine kinase inhibitors (TKIs) have challenged this paradigm due to high intracranial response rates in patient subgroups. Drug-conjugated antibodies (ADCs), such as trastuzumab deruxtecan, represent a new class of compounds increasingly used in systemic cancer treatment. This review aims to analyze the efficacy of ADCs in treating primary and secondary brain tumors, while exploring strategies to enhance their effectiveness. These findings highlight strategies to enhance ADCs’ intracranial efficacy and the need to refine the design of clinical trials including patients with intracranial untreated metastases.

Abstract

Primary and secondary brain tumors have always been a challenge due to their high morbidity and poor prognosis. The incidence of brain metastasis is also increasing with the advent of effective new treatments. Traditional systemic treatments have historically had limited success, partly due to poor central nervous system (CNS) penetration. However, the advent in recent decades of new therapies that have shown high encephalic response rates are challenging this paradigm. ADCs represent a new class of compounds revolutionizing cancer treatment with high systemic response rates and lower toxicities. The continuing evolution of ADCs has shown that certain structural features such as payload, linker, and drug-to-antibody ratio (DAR) are essential in determining their efficacy at the encephalic level, and some ADCs have started to exhibit promising efficacy in treating primary and secondary brain tumors. Unfortunately, most patients with untreated encephalic metastases are excluded from clinical trials, with data primarily from retrospective studies or post hoc analyses. This review describes the early signs of ADC efficacy in brain tumors, the role of complementary treatments like radiation therapy, and critical points to improve ADC efficacy in brain malignancies.

1. Introduction

Primary and secondary brain tumors still remain a significant challenge in modern oncology due to their high morbidity and poor prognosis [1,2].
The overall annual cumulative incidence of primary malignant CNS tumors in the US is approximately 6.94 per 100,000 person-years [3]. Among them, glioblastoma (GBM) represents the most common and deadly form in adults, with a 5-year overall survival (OS) rate of 7.2% despite the aggressive multidisciplinary approach used to treat it, including surgery, radiotherapy (RT) and chemotherapy [4].
Brain metastases (BMs) are ten times more common than primary brain tumors (pBTs) [5], with approximately 20% of patients with advanced solid tumors developing BMs during their lifetime, particularly those with lung and breast cancers and melanoma [6].
Despite the use of surgery, RT, and chemotherapy, and the recent advent of novel effective agents such as immunotherapy and targeted therapy, the prognosis of patients with BMs remains generally poor [7]. Moreover, CNS malignancies are often highly symptomatic, with a devastating impact on patients’ quality of life, further compromised by the potential side effects of radiation and surgical treatments [8,9].
Currently, BMs are primarily managed with local treatments such as RT and, in selected cases, surgery. Radiotherapy approaches have evolved over time, transitioning from traditional whole-brain RT to more accurate stereotactic radiosurgery, leading to better disease control while minimizing neurocognitive side effects [10].
Historically, the effectiveness of systemic treatment for CNS tumors has been limited by the presence of the blood–brain barrier (BBB) [11], which is responsible for maintaining the homeostasis of CNS but often impedes adequate drug penetration and the bioavailability of the most used anticancer agents.
The BBB consists of a layer of endothelial cells that surround the blood vessels, supported by cells such as astrocytes and pericytes. Endothelial cells encircling vessels in the CNS differ from peripheral endothelial cells, as they lack fenestrations and are bound by both tight junctions and adherens junctions that prevent molecules from entering the encephalic circulation. The BBB is equipped with a series of transport systems that facilitate the entry of nutrients and other substances. Small (<400–500 Da) and highly lipophilic molecules can passively cross the BBB thanks to their physicochemical properties, while molecules >550 Da such as monoclonal antibodies (mAbs) need energy-dependent transport systems such as the receptor-mediated transcytosis (RMT). Active efflux mechanisms at the BBB level complicate the CNS penetrance of various compounds even further. Among these, the most well known is P glycoprotein (P-gp), an ATP-dependent efflux transporter highly expressed at the BBB, the function of which is to expel harmful substances from the CNS circulation, including several chemotherapeutic agents, which further limits their accumulation in the CNS. [12]
However, in the presence of growing tumors, the BBB is often altered and disrupted; on the other hand, the blood–tumor barrier (BTB) [13], which is highly heterogeneous but generally more permeable than the BBB, allows larger molecules to traffic into CNS parenchyma [11,14,15].
High-grade gliomas often exhibit a dysfunctional BTB in the central regions, leading to contrast enhancement on T1-weighted MRI sequences, while the peripheral regions, where the tumor exhibits infiltrative growth, maintain an intact BBB. Compared to pBTs, metastatic lesions typically show a more consistent disruption of the BBB and, generally, no infiltrative growth, which facilitates diagnostic imaging and, when feasible, surgical resection. The suboptimal and highly variable drug accumulation in brain tumors mainly depends on the BTB’s heterogeneous permeability for small and large molecules as well as on the heterogeneous perfusion.
The new wave of anticancer agents developed over the last two decades, such as TKIs targeting tumor driver genetic alterations and immune checkpoint inhibitors (ICI), which have revolutionized the treatment paradigm of most cancers recently, has exhibited an unprecedented level of activity in the CNS. Currently the effectiveness of these compounds challenges, at least in some clinical scenarios, the old paradigm of BM treatment management based on local interventions.
ADCs represent an innovative class of antineoplastic drugs that allow the selective delivery of cytotoxics into tumor cells [16], and they are quickly emerging as part of a major breakthrough in cancer therapy in recent years.
As of now, there are only seven ADCs that have received FDA approval for the treatment of solid tumors (Table 1).
An ADC consists of three main components: the antibody, the linker, and the payload. Linkers can be distinguished into cleavable and non-cleavable linkers. In the case of cleavable linkers, the antibody binds to a target molecule on the tumor cell’s surface, at which point the linker is degraded by conditions in the tumor microenvironment, leading to internalization of the drug. Non-cleavable linkers, in contrast, require endocytosis and degradation of the entire ADC structure to release the payload inside the cell. The resulting cell death enables the payload to affect nearby tumor cells, even those not expressing the target, through a mechanism called the bystander effect (Figure 1) [17].
Non-cleavable linkers are more stable in the bloodstream, but due to their lower cell permeability, they limit the bystander effect, which is crucial when dealing with the heterogeneous expression of the targeted antigen within the tumor [18].
The structural characteristics of an ADC can drastically change the activity of the compound, leading to different levels of efficacy between antibodies, even in the presence of the same target [19].
A crucial factor that determines the effectiveness of ADCs is the DAR, which is the number of chemotherapy molecules carried per single antibody. The DAR can affect ADCs’ stability, solubility, and pharmacodynamics, resulting in different efficacies. A higher DAR might result in better cytotoxicity and an increased bystander effect but lead to more rapid clearance.
Molecules as large as ADCs traditionally cannot cross an intact blood–brain barrier.
However, this paradigm is being challenged by the new generation of ADCs, which have demonstrated high rates of intracranial activity, especially in lung and breast cancer, thus suggesting good brain penetrance. However, the same level of intracranial activity was not observed in the context of primary brain tumors: depatuxizumab mafodotin (Depatux-M), despite its promises, has failed to demonstrate efficacy in patients with EGFR-amplified GBM in both the adjuvant and recurrent treatment settings. In 2021, preclinical studies by Marin et al. showed how the inefficacy of Depatux-M is likely due to insufficient uptake at the encephalic level [20]. This difference in efficacy may be explained by both the differing vascularization and destruction of the BBB between pBTs and BMs.
In addition to intrinsic factors of the BBB, structural features of ADCs, including homogeneity, hydrophobicity, DAR and payload type, seem to be key determinants in the encephalic distribution.
In this comprehensive review, we will thoroughly explore and analyze current evidence on the intracranial activity of ADCs in pBTs and BMs from different primary cancers, including only those tumor types for which there is in a sufficient amount of data in the literature. Despite the current lack of evidence supporting the efficacy of ADCs in the treatment of pBTs, we included a section about the different trials that have been conducted in this setting, from the first attempts with immunotoxins, to the latest INTELLANCE-1 trial, the only phase III study published in this context as of today.
We will further discuss efforts to increase ADC delivery into the brain, the role of radiotherapy as well as biomarkers that could guide us in selecting the optimal treatment for our patients.
The study selection process highlighted that, beyond the inherent biological complexities, substantial methodological challenges persist in designing high-quality systemic therapy trials for patients with brain metastases (BMs) and primary brain tumors (pBTs). These challenges include modernizing eligibility criteria, the use of suboptimal CNS-specific endpoints, inadequate consideration of efficacy denominators and prior radiation exposure, inconsistent application of disease measurability criteria (e.g., RANO, RECIST), and persistent confusion regarding the capture and differentiation of CNS-specific versus extra-CNS efficacy outcomes.
To our knowledge, this is the first narrative review to assess the currently available data on the efficacy of ADCs in both BMs and pBTs and to explore possible strategies to enhance therapeutic effectiveness, including pharmacologic optimization and combination with radiotherapy, with the aim of guiding clinicians toward the most appropriate treatment choices for their patients
Approximately 30 articles presenting clinical trial results published globally between 2007 and 2024 were identified through searches conducted in PubMed, ClinicalTrials.gov, Scopus, and the Cochrane Library. The search utilized keywords including glioblastoma, central nervous system, brain metastases, antibody–drug conjugate, linker, blood-brain-barrier, biomarkers, liquid biopsy and their combinations. Titles and abstracts were screened for relevance to this study. Additionally, the reference lists of the selected reviews and articles were examined to identify further relevant sources.

2. Primary Brain Tumors

Given the dismal outcomes and the urgent need for more effective therapeutic options, ADCs have been tested for their potential in treating gliomas.
Immunotoxins could be considered the first form of ADCs designed for GBM treatment. These compounds are constituted of toxins derived from bacteria or plants, conjugated to an antibody targeting a tumor antigen. Pseudomonas aeruginosa exotoxin A (PE) and Diphtheria toxin (DT) are two of the most used toxins in this setting due to their ability to inhibit protein synthesis by modifying elongation factor-2 or by direct ribosomal inhibition [21,22]. These agents’ efficacy might be provided by their specificity, potency, immunity to resistance mechanisms, and independence on cell cycle kinetics [23].
The first immunotoxins developed for GBM targeted the epidermal growth factor receptor (EGFR) pathway [24].
The epidermal growth factor receptor variant III (EGFRvIII) is the most common EGFR mutation occurring in approximately 25–64% of GBM cases; it derives from the in-frame deletion of exons 2–7 of the EGFR gene and codes for a constitutively active, oncogenic, ligand-independent receptor. EGFR amplification is also common in GBM, being present in ~50% of cases [25] (35% in the Asian population) [26]. EGFR amplifications co-exist with EGFRvIII in ~50% of cases (~25% of GBM overall) [27].
TP-38 is a recombinant chimeric protein containing a genetically engineered form of the PE whose native binding domain has been replaced with transforming growth factor-α (TGF-α), making it able to specifically target EGFR [23]. A phase I study of TP-38 administered to patients with progressive or recurrent primary or metastatic brain tumors (including 17 patients with GBM, 1 with gliosarcoma, 1 with anaplastic oligodendroglioma and 1 patient with spindle cell metastasis) through an intracerebral convection-enhanced delivery (CED) technique has been conducted. This study closed prematurely during the dose escalation phase due to inconsistent drug delivery. The median survival of the overall population was 28 weeks (95% CI, 26.5–102.8), while of 15 patients with residual measurable disease, 2 (13.3%) had radiographic responses, including 1 GBM patient, who had a nearly complete response (CR) [23].
Beyond EGFR, other targets being investigated included interleukin-13 (IL-13), interleukin-4 (IL-4) and the transferrin receptor (Table 2).
A randomized phase III study compared the efficacy of Cintredekin besudotox (CB), an immunotoxin composed of IL-13 and an engineered truncated form of PE delivered intracranially through CED, with Gliadel wafers as a form of treatment of GBM at first recurrence, after a salvage surgical resection. No survival difference was observed between the two treatment arms [28] and the safety profiles were comparable, except for the number of pulmonary embolisms, which was higher in patients treated with CB.
MDNA55 (Bizaxofusp), an IL-4 receptor-targeting toxin administered intratumorally via CED, has been investigated as a single agent in a single-arm phase IIb study including 47 patients with recurrent GBM (rGBM). MDNA55 showed an acceptable safety profile at doses up to 240 μg. The overall tumor control rate evaluated according to modified Response Assessment in Neuro-Oncology (RANO) criteria, including tumor shrinkage or stabilization, was 75.6% (31 of 41 patients), with 1 patient exhibiting a durable CR following initial radiographic changes consistent with pseudoprogression. The most significant benefit has been observed in the subgroup of IL4R High + IL4R LowHD patients with a TCR of 81%. The median overall survival (mOS) and OS at 12 months were 11.64 months (80% one-sided CI 8.62, 15.02) and 46%, respectively [29]. Based on these results, it has been since granted Fast Track (Food and Drug Administration—FDA) and orphan drug status (FDA and European Medicines Agency—EMA) for the treatment of rGBM [30].
Tf-CRM107 is an immunotoxin carrying a modified DT payload covalently bound to transferrin [31]. Two phase III trials of Tf-CRM107 were initiated, but unfortunately, one was withdrawn following a probability analysis performed prior to the recruitment of patients, determining that it was unlikely that the agent would meet FDA criteria for efficacy (NCT00083447; clinicaltrials.gov, Accessed on 5 May 2025). The other one instead compared Tf-CRM107 with standard treatment. Despite the accrual having been completed, results were never released (NCT00088400; clinicaltrials.gov, Accessed on 5 May 2025).
More recently, the ADC that raised the greatest expectations for GBM treatment was Depatux-M. This agent comprises an anti-EGFRvIII mAb (depatuximab) connected via a non-cleavable linker to a microtubule inhibitor, monomethylauristatin-F (MMAF). The mAb depatuximab not only targets the EGFRvIII, but is also able to bind to wild-type EGFR on neoplastic cells with EGFR amplification, even in the absence of EGFRvIII co-mutation, by binding to a tumor-specific, conformationally exposed epitope [32].
The safety, pharmacokinetics (PK), and preliminary antitumor activity of Depatux-M, given either alone in rGBM or in combination with temozolomide (TMZ) in newly diagnosed (nGBM) or rGBM, were tested in a large multicenter, three-arm, phase I trial. Arm A included patients with nGBM who were to receive Depatux-M + RT + TMZ; arm B included both newly diagnosed or recurrent GBM patients who were to receive Depatux-M + TMZ after RT; and arm C included enrolled patients with rGBM who were to receive Depatux-M in monotherapy. Each arm was composed of a dose escalation and dose expansion cohort. The most common adverse events (AEs) were ocular, occurring in 87% of patients, including blurred vision (63%) and photophobia (35%), while the most common non-ocular AEs were thrombocytopenia (45%, attributable to TMZ, potentially exacerbated by concurrent Depatux-m) and fatigue (38%). Dose delays occurred in 35 patients (58%), most commonly due to ocular side effects, while no treatment discontinuations due to ocular side effects were reported. PK exposures of Depatux-M, total Depatux, and cys-mafodotin after i.v. infusion of Depatux-M were approximately dose-proportional over the dose range of 0.5–1.5 mg/kg. Coadministration of Depatux-M did not affect the PK parameters of TMZ. Among 58 patients with RANO-defined measurable disease at baseline, the overall response rate (ORR) was 13.8% (1/58 CR, 7/58 PR, 95% CI = 6.2%, 25.4%), 26 patients had stable disease (SD), and 24 patients had progressive disease (PD). The median duration of response (mDOR) was 5.6 months (95% CI = 1.5, 9.7). The median progression-free survival (mPFS) and mOS of the entire study population were 2.1 months (95% CI = 1.1, 3.4) and 7.4 months (95% CI = 6.5, 9.6), respectively.
Based on this acceptable safety profile and the preliminary efficacy results, Depatux-M [33,34,35,36] was further tested in larger phase II and III trials.
Table 2. Trials on immunotoxins with targets other than EGFR in GBM.
Table 2. Trials on immunotoxins with targets other than EGFR in GBM.
Reference Type of Trial Population Intervention Target Endpoints
[35] Phase III rGBM
(first recurrence)
CB vs. gliadel wafers IL-13 No survival difference
[36] Phase IIb rGBM MDNA55 (Bizaxofusp) IL-4 mOS: 11.64 months [80% one-sided CI 8.62, 15.02]
Abbreviations: CB, Cintredekin besudotox; rGBM, recurrent GBM.
In the randomized phase II study INTELLANCE-2, 260 patients with EGFR-amplified rGBM were randomized to receive Depatux-M in combination with TMZ (ARM 1), Depatux-M alone (ARM 2), TMZ or lomustine (ARM 3). At a median follow-up of 28.7 months, a significant benefit in OS was observed in the combination arm vs. control arm 3 (HR 0.66, 95% CI 0.47–0.93; p = 0.017). Conversely, no significant difference was observed between the Depatux-M monotherapy and the control arm (HR 0.96, 95% CI 0.69–1.33; p = 0.80). The 2-year survival was 19.8% (95% CI 12.2, 28.8), 10% (95% CI 4.8, 17.6), and 5.2% (95% CI 1.7, 11.7), in the combination arm, Depatux-M monotherapy arm, and control arm, respectively. No significant differences according to the O6-Methylguanine-DNA methyltransferase (MGMT) promoter methylation status were observed, neither in the monotherapy nor in the combination arms. A trend toward increased clinical benefit from the combination of Depatux-M + TMZ was seen in patients relapsing > 16 weeks after the end of TMZ treatment, with a 2-year survival of 28.6% (95% CI 13.5, 45.6) for the combination arm, vs. 11.1% (95% CI 2.8, 25.9) for the Depatux-M monotherapy arm, and 3.9% (95% CI 0.3, 16.4) for the control arm [37].
The subsequent INTELLANCE-1 study was a large randomized, placebo-controlled phase III trial designed to compare the addition of Depatux-M to chemo-radiation vs. the standard Stupp regimen, in patients with EGFR-amplified nGBM, with OS as the primary endpoint. The trial did not meet its primary endpoint, showing no OS benefit for the experimental arm with an mOS of 18.9 vs. 18.7 months in the control arm (HR 1.02, 95% CI 0.82–1.26; 1-sided p = 0.63). Progression-free survival (PFS) was longer for patients who received Depatux-M compared to those who received a placebo, with an mPFS of 8.0 vs. 6.3 months (HR 0.84, 95% CI 0.70–1.01; p = 0.029). This benefit was more relevant in patients harboring the EGFRvIII mutation (mPFS 8.3 vs. 5.9 months, HR 0.72, 95% CI 0.56–0.93; 1-sided p = 0.002) and for MGMT-unmethylated patients (HR 0.77, 95% CI 0.61–0.97; 1-sided p = 0.012). These data raised the question of whether the EGFR-vIII mutation might represent a better selection biomarker for trial eligibility rather than amplification. The enrolled population in this case included only approximately 50% of EGFRvIII-mutated patients. However, it is important to underscore that high intratumor heterogeneity significantly impacts the predictive value of both EGFRvIII and receptor amplification [38]. Corneal epitheliopathy was confirmed as the most common treatment-related adverse event (TRAE) occurring in 94% of patients of the experimental arm, of which 61% were grade 3–4, leading to a 12% discontinuation rate.
Given the failure to meet the primary endpoint, this study was discontinued early and unblinded for futility [39].
A real-world study of Depatux-M + TMZ in rGBM was conducted by an Italian group and demonstrated results comparable with those of the phase II trial INTELLANCE-2, finding that the only factor significantly associated with a better outcome was the MGMT methylation status [40].
Another transmembrane protein, the human trophoblast cell surface antigen 2 (TROP2), was recently correlated with GBM proliferation capacity [41]. Interestingly, its amplification was associated with its promoter hypomethylation, albeit in a very small sample [42,43].
Moreover, normal human brain tissue does not express TROP2, potentially increasing the therapeutic index of ADCs in this setting [44]. A phase II multicenter, prospective study of SG in rGBM has now entered the interim analyses [41].
Currently ongoing trials assessing the efficacy of ADCs in primary CNS tumors are listed in Table 3.

3. Brain Metastases

3.1. Non-Small Cell Lung Cancer

Approximately 50% of patients with advanced non-small cell lung cancer (NSCLC) develop BMs throughout the course of their disease. The incidence of BMs is particularly high in NSCLC, with EGFR mutations or anaplastic lymphoma kinase (ALK) rearrangements with a baseline occurrence around of 30–35% in both groups [45,46].
The emergence of BMs represents a relevant challenge in the clinical management of these patients, given their association with poor prognosis, neurological deterioration, and reduced quality of life.
As underlined above, the historical management of BMs, relying on RT and surgery, has been recently challenged by the advent of new anticancer agents, such as ICIs and TKIs targeting actionable genetic alterations. This is particularly true in the context of lung cancer, with the entry into the market of several compounds which have proven to be truly active in the CNS, leading to excellent intracranial disease control [47]; this is the case for EGFR and ALK inhibitors or anti PD-(L)1 monoclonal antibodies (mABs).
In a subgroup analysis of the phase III FLAURA trial, osimertinib, a third-generation EGFR TKI, compared to erlotinib or gefitinib in a population of treatment-naïve, EGFRm NSCLC patients, was associated with a 52% reduction in the risk of CNS progression or death due to its superior ability to cross the BBB [48]. The great ability of osimertinib in controlling intracranial disease was further confirmed in the phase III FLAURA 2, with an intracranial objective response rate (iORR) of 73% being reached in the chemotherapy combination arm and 69% in the monotherapy arm [49].
In a population of treatment-naïve ALK-positive NSCLC patients with BMs enrolled in a phase III CROWN study, the third-generation ALK TKI lorlatinib led to an impressive iORR, assessed by a RECIST v1.1 value of 60%, with an intracranial complete response rate (iCRR) of 49% [50].
In non-oncogene-addicted metastatic NSCLC, a pooled analysis of the KEYNOTE-021, KEYNOTE-189, and KEYNOTE-407 studies, which included patients with stable BMs at baseline, showed that the addition of pembrolizumab significantly improved OS compared to chemotherapy alone. Unfortunately, iORRs are not available since the lesions were considered to be non-targets within the studies [51].
In the ATEZO-Brain [52] trial, the combination of anti PD-L1 Atezolizumab + chemotherapy in a population of NSCLC patients and untreated BM achieved an iORR of 42.7%, assessed with RANO and RECIST 1.1 criteria. In the CAP-BRAIN [53] trial, Camrelizumab + chemotherapy was evaluated in a population, with more than three MRI-confirmed BMs or one to two BMs not amenable for local therapy. Camrelizumab plus chemotherapy led to an iORR of 46.7%, as assessed with RECIST 1.1 criteria.
In this quickly evolving landscape, ADCs are emerging as a new therapeutic strategy for the treatment of NSCLC, and understanding their degree of CNS activity is crucial to determining if they represent a novel option even in the presence of BMs, particularly when RT is not feasible, chemo-immunotherapy fails and resistance to targeted drugs occurs [54].

3.1.1. Trastuzumab Deruxtecan

To date, the first and only FDA-approved ADC for the treatment of lung cancer is trastuzumab deruxtecan (T-DXd). T-DXd consists of an antibody targeting the human epidermal growth factor receptor 2 (HER2) linked to a topoisomerase I inhibitor, cytotoxic payload deruxtecan (DX-d), via a cleavable linker. HER2 is a transmembrane receptor of the EGFR family that activates through dimerization, promoting HER2 pathway activation. In NSCLC, up to 20% of patients have HER2 overexpression while only 2% bear a targetable mutation. These mutations are commonly linked to adenocarcinoma histology and are more frequently observed in females, individuals of Asian descent, and those who have never smoked [55].
In August 2022, the FDA granted accelerated approval of T-DXd for the treatment of unresectable/metastatic NSCLC patients with an HER2 mutation, based on results of the DESTINY-Lung01 and DESTINY-Lung02 trials.
The DESTINY-Lung01 and DESTINY-Lung02 trials were multicenter, single-arm, phase II studies evaluating the efficacy of T-DXd in patients with unresectable or metastatic NSCLC carrying HER2 mutations or overexpression. DESTINY-Lung01 enrolled a total of 181 patients; 91 patients were enrolled in cohort 2 (HER2 mutated cohort) and treated with T-DXd at a dose of 6.4 mg/kg every 3 weeks, while 90 patients were enrolled in cohort 1 (HER2 overexpressing cohort) and treated with an either 5.4 mg/Kg (cohort 1) or 6.4 mg/kg dose (cohort 1a). DESTINY-Lung02 enrolled 152 patients with HER2 mutation who were randomized to receive either T-DXd at a dose of 5.4 mg/kg or 6.4 mg/kg every 3 weeks. Patients with stable (asymptomatic and not requiring steroid treatment) BMs were allowed to participate in both studies. In the pooled analyses from DESTINY-Lung01 and DESTINY-Lung02 trials, T-DXd showed excellent intracranial activity. In the 5.4 mg/kg treatment cohort, the iORR, according to RECIST v1.1, was 25% (8/32), with four iCRs and four intracranial partial responses (iPRs), while an intracranial stable disease (iSD) was observed in 56.3% (18/32) of cases. In the 6.4 mg/kg cohort, 10 out of 54 (18.5%) patients had a response to T-DXd, including 9 iPRs and 1 iCRs, while 31 had patients had an iSD (57.4%) [56].
Nevertheless, many ADCs are currently under development for the treatment of NSCLC. In this section, we will review the key ADCs being investigated and those for which intracranial response data are available (Table 4).

3.1.2. Datopotamab Deruxtecan

Datopotamab deruxtecan (Dato-DXd) is an ADC that targets the TROP2 antigen, linked to the topoisomerase I inhibitor deruxtecan via a cleavable linker. Trop2 is a type-1 surface glycoprotein that binds to various partners such as claudins 1 and 7, cyclin D1, and insulin-like growth factor 1 (IGF-1) [57]. It plays an important role in stabilizing epithelial tight junctions and regulating cell growth and proliferation, and is overexpressed in approximately 64% of adenocarcinomas and up to 75% of squamous cell carcinomas [58,59]. In the single-arm phase II TROPION-Lung05 trial, Dato-DXd was investigated as a treatment for patients with metastatic NSCLC and at least one or more genomic alterations, including EGFR, ALK, ROS proto oncogene 1 (ROS1), neurotrophic tyrosine receptor kinase (NTRK), b-raf proto-oncogene (BRAF), rearranged during transfection (RET), or mesenchymal–epithelial transition factor (MET) exon 14 skipping, who received up to four prior lines of treatment, including at least one target therapy. Patients with BMs were eligible if they were asymptomatic without steroid therapy.
Data on intracranial activity from a prespecified exploratory analysis were presented at the 2024 ASCO annual meeting. Among the 53 patients with BMs, treatment with Dato-DXd led to an intriguing iORR of 22%, as assessed by RECIST v1.1, along with an intracranial progression-free survival (iPFS) of 6.9 months. The ORR was 28% in patients with baseline BMs and 40% in those without.

3.1.3. Patritumab Deruxtecan

Patritumab deruxtecan (HER3-DXd) is an ADC that targets the human epidermal growth factor 3 (HER3) antigen, linked to the topoisomerase I inhibitor DXd via a cleavable linker. HER3 is often overexpressed in many tumors, including NSCLC, with a prevalence of 83% in primary tumors. Binding to its ligand, heregulin, HER-3 increases tumor proliferation and metastatic capacity. Recently, the role of HER3 as a resistance factor to TKIs has emerged. Dimerization with other receptors such as HER2, EGFR, and MET leads to proliferation signals that may cause cells to resist targeted therapy [60,61]. HER3-Dxd has been evaluated in the phase II HERTHENA-Lung01 (NCT04619004) trial in participants with metastatic or locally advanced NSCLC with canonical activating EGFR mutations (exon 19 deletion or L858R) who have received and progressed on or after at least one EGFR TKI and one platinum-based chemotherapy-containing regimen. Patients with baseline BMs were eligible if clinically silent with a need of daily prednisone <10 mg/day or equivalent. Out of 225 total patients, 30 had non-irradiated BM at baseline. In this subgroup, HER3-DXd demonstrated an iORR of 33.3% as assessed by Blinded Independent Central Review (BICR) per RECIST criteria.

3.1.4. Telisotuzumab–Vedotin

Telisotuzumab–Vedotin (Teliso-V) is a c-MET-directed ADC with a monomethyl auristatin E cytotoxic payload. Met is a proto-oncogene that is activated by hepatocyte growth factor (HGF). Its hyperactivation in NSCLC is mediated by several mechanisms such as amplification and skipping of exon 14 [62].
The Phase 2 LUMINOSITY trial evaluated Teliso-V in locally advanced/metastatic non-squamous NSCLC patients with MET overexpression. Patients with BMs were eligible if the metastases received definitive treatment and were stable. Out of the 161 NSCLC patients included in the study, 33 (20.5%) had stable and previously treated BMs at the study entry. The trial demonstrated an ORR of 34.6% in c-Met high patients, 22.6% in c-Met intermediate patients and 28.6% in the intention to treat (ITT) population. Unfortunately, subgroup analyses regarding intracranial activity have not yet been presented [63].
Several other ADCs targeting molecules such as EGFR, folate receptor 1 (FOLR1), receptor tyrosine kinase-like orphan receptor 2 (ROR2) and AXL receptor tyrosine kinase (AXL) are currently under investigation for the treatment of NSCLC, but data on intracranial activity are not yet available (Table A1).

3.2. Breast Cancer

Breast cancer (BC) is the second most common malignancy causing BMs, after lung cancer [64]. Among the BC molecular subgroups, HER2-positive and triple-negative breast cancer (TNBC) are those most frequently associated with the development of BMs, with a cumulative incidence of 31% and 32%, respectively, while CNS involvement is rarer in the luminal subtype, with a cumulative incidence of 15% [65].
The prognosis of patients with BC and BMs remains quite poor, with an mOS of 8.7 months, but there are significant differences according to the molecular subtypes, ranging from 9.3, 16.5, and 21.6 to 4.9 months in the luminal, HER2-positive and TNBC group, respectively [66].
Recently, Chehade et al. demonstrated that most surgically resected BMs can be classified as HER-2-low by immunohistochemistry (IHC), thus potentially being druggable with HER-2-directed ADCs [67]. Another potential target was highlighted in a phase 0 trial, where patients with breast cancer brain metastasis (BCBM) received SG before surgery. In this study, 100% of TNBC BM tumor samples demonstrated TROP2 expression with a 3+ score for IHC [68].
In this section, we will review FDA-approved ADCs for treating BC, for which intracranial response data are available, including Trastuzumab emtansine (T-DM1), T-DXd and Sacituzumab Govitecan (SG) (Table 5).
T-DM1 comprises the mAb trastuzumab conjugated via a non-cleavable linker to the potent tubulin polymerisation inhibitor mertansine (DM1). It is indicated in the adjuvant treatment of patients with HER2-positive early breast cancer with residual invasive disease after the administration of a trastuzumab- and taxane-based neoadjuvant regimen and in the advanced setting, for patients with HER2-positive MBC who previously received trastuzumab and a taxane, separately or in combination.
T-DXd is indicated for the treatment of HER2-positive advanced breast cancer (aBC), previously treated with an anti-HER2-based regimen in the metastatic/neoadjuvant/adjuvant setting, if disease recurrence occurs during or within 6 months of completing therapy. In August 2022, the FDA approved T-DXd also for the treatment of patients with unresectable or metastatic HER2-low (immunohistochemistry [IHC] IHC 1+ or IHC 2+ with a negative in situ hybridization test [ISH−]) breast cancer, who had received prior chemotherapy in the metastatic setting or developed disease recurrence during or within 6 months of completing adjuvant chemotherapy.
The third approved ADC in the context of aBC is SG, a conjugate composed of an antibody targeting the antigen TROP2, which is expressed in most breast cancers, coupled to SN-38 (topoisomerase I inhibitor) through a hydrolyzable linker. SG was approved by the FDA on February 2023 for patients with unresectable, locally advanced or metastatic hormone receptor (HR)-positive, HER2-negative (IHC 0, IHC 1+, or IHC 2+ with ISH−]) breast cancer who have received endocrine-based therapy and at least two additional systemic therapies in the metastatic setting.

3.2.1. Ado-Trastuzumab Emtansine

In the (neo)adjuvant setting, T-DM1 did not demonstrate a decrease in the risk of BMs, probably because of its scarce penetration into an intact BBB [69,70].
The randomized phase III EMILIA trial established T-DM1 as the standard second-line treatment for patients with HER2-positive aBC previously treated with trastuzumab and a taxane, showing superiority over lapatinib + capecitabine (XL) in terms of PFS and OS (mOS of 29.9 months (95% CI 26.3–34.1) vs. 25.9 (95% CI 22.7–28.3); HR 0.75 (95% CI 0.64–0.88) [71].
A retrospective exploratory analysis of this trial assessed the incidence of BMs after treatment with T-DM1 vs. XL and treatment efficacy among patients with pre-existing CNS metastases. Considering patients with no BMs at baseline, CNS progression occurred in 9 of 450 (2%) in the T-DM1 and in 3 of 446 (0.7%) in the XL arms. Among the 95 patients with previously treated and asymptomatic BMs at baseline, T-DM1 (n = 45) has been demonstrated to be superior to XL in prolonging intracranial disease control in this specific population, with a mPFS of 5.9 months (vs 5.7 in the control arm) (HR 1.00; 95% CI 0.54–1.84; p = 1.000), and an mOS of 26.8 (vs 12.9 months in the control arm) (HR 0.38, 95% CI 0.18–0.79; p = 0.0081) [72].
In the randomized, open-label, phase III TH3RESA trial, T-DM1 was compared to the treatment of physician’s choice (TPC) for patients with metastatic breast cancer (mBC) pretreated with ≥2 HER2-directed regimens. The systemic mPFS was 6.2 vs. 3.3 months in the T-DM1 group vs. the control arm, respectively (HR 0.53, 95% CI 0.422–0.661; p < 0.0001). Considering only patients with asymptomatic, previously treated CNS metastases, the mPFS was 5.8 months in the T-DM1 (n = 40) arm vs. 2.9 months in the control arm (n = 27) (HR 0.47, 95% CI; 0.24–0.89) [73].
The single-arm, open-label phase IIIb KAMILLA trial [74] tested T-DM1 in 2002 patients with HER2-positive locally a/mBC treated with prior HER2-targeted therapy and chemotherapy, of which 398 had BMs (controlled or untreated and asymptomatic). Tumor response and clinical outcomes in patients with baseline BMs were evaluated in a post hoc exploratory analysis. Among the 126 cases with measurable BMs, according to RECIST v1.1, the iORR was 21.4%, with an iCR of 2.4%. The mPFS was 5.5 months (95% CI 5.3–5.6) and the mOS was 18.9 months (95% CI 17.1–21.3). A reduction of 30% or more in the sum of the largest diameters of target brain lesions was observed in 50.0% (5/10; 95% CI 18.7–81.3) and 32.7% (16/49; 95% CI 20.0–47.6) of patients treated with brain RT < 30 days and ≥30 days before study entry, respectively, and in 49.3% of those who did not receive brain RT (33/67; 95% CI 36.9–61.8).

3.2.2. Trastuzumab Deruxtecan

Another agent recently introduced among the therapeutic armamentarium of HER2-positive mBC is T-DXd.
In the phase II DESTINY-Breast01 trial, 24 patients with previously treated and asymptomatic BMs were included. The iORR, according to RECIST v1.1 criteria, of the 17 patients with measurable BMs at baseline was 41.2% [75,76], while the mPFS of the entire BM cohort was 18.1 months (95% CI 6.7–18.1).
The phase III DESTINY-Breast03 trial evaluated the efficacy and safety of T-DXd vs. T-DM1 in HER2-positive mBC patients who progressed to trastuzumab and a taxane. In the 82 patients with inactive and/or previously treated BMs enrolled in this trial, T-DXd showed a significantly improved iORR, according to RECIST v1.1 criteria, compared to T-DM1 (79 vs. 35%, respectively) [19]. The considerable CNS activity of T-DXd was then confirmed in the TUXEDO-1 trial, a prospective, single-arm, phase 2 study recruiting patients with HER2-positive BC and untreated BMs or BMs progressing after previous local therapy, after previous exposure to trastuzumab and pertuzumab and with no indication for immediate local therapy. Among the 15 patients enrolled in the ITT population, the iORR was 73.3% (95% CI 48.1–89.1%), meeting the predefined primary endpoint, with 2 patients (13.3%) experiencing an iCR, 9 (60%) an iPR and 3 (20%) iSD [77].
The largest amount of prospective data of T-DXd in patients with BMs so far have come from the DESTINY-Breast12 trial [78]. This is a multicenter, phase IIIb/IV, open-label, two-cohort study of T-DXd in patients with previously treated HER2-positive mBC with and without BMs. Of the 504 patients enrolled in the trial, 263 had BMs, of which 157 were previously treated and considered stable, while 106 were active (39 untreated and 67 previously treated BMs progressive at time of study entry—without indication for immediate retreatment with local therapy). T-DXd demonstrated substantial and durable intracranial clinical activity, with an overall CNS response rate of 71.7% within the 138 cases with baseline measurable CNS disease (stable BMs: n = 77; active BMs: n = 61). The iORR was 79.2% (95% CI 70.2–88.3) and 62.3% (95% CI 50.1–74.5) in patients with stable and active BMs, respectively. Interestingly, the 12-month survival rate of patients with and without BMs was comparable, at 90.6% and 90.3%, respectively. The safety profile of T-DXd was consistent with that in previous reports, while interstitial lung disease/pneumonitis was confirmed as an important safety risk of T-DXd, with six related deaths in the BM cohort, five of which occurred in patients taking steroids.
Another example of strong confirmation of T-DXd CNS activity can be found in the ongoing phase II DEBBRAH trial. This study evaluates patients with pretreated HER2-positive or HER2-low advanced BC with stable, untreated, or progressing BMs, and/or leptomeningeal carcinomatosis, allocated into five cohorts based on HER2 expression and metastatic brain status. Cohort 1 comprises patients with non-progressing BMs after local therapy (n = 8), cohort 2 includes asymptomatic untreated BMs (n = 4), and cohort 3 includes those with progressing BMs after local therapy (n = 9). Overall, the iORR in patients with active BMs was 46.2% (95% CI, 19.2–74.9), 50.0% (95% CI, 6.7–93.2) in cohort 2 and 44.4% (95% CI, 13.7–78.8; p < 0.001) in cohort 3, respectively. In cohort 1, the 16-week PFS rate was 87.5% (95% CI, 47.3–99.7; p < 0.001) [79].
Currently, the evidence on HER2-low BCBM is limited. A deeper understanding of the clinicopathological characteristics and prognosis of HER2-low BCBM is essential. The results attained by T-DXd in HER-2 low and HER-2 ultra-low subtypes leverage a definition as a spectrum continuum of expression of HER-2 in mBC [80]. The phase 2 DAISY trial evaluated the T-DXd response rate (RR) based on HER-2 expression, including those with treated and asymptomatic BM [81,82]. Results showed an ORR of 70%, 36%, and 30% for HER-2 positive (3+ by IHC), HER-2 low (1+ or 2+ by IHC, ISH negative), and HER-2 negative (0+ by IHC) BC cohorts, respectively. Similarly, in the DESTINY-Breast04 trial, 35 patients with stable and treated BMs showed improved intracranial responses with T-DXd in HER2-low disease compared to the TPC [83]. Results from cohorts 2 and 4 of the DEBBRAH trial evaluating T-DXd in patients with active HER2-low BMs showed an iORR per RANO-BM of 50.0% in patients with asymptomatic untreated BMs (n = 6, cohort 2) and 33.3% in those with progressing BMs (n = 6, cohort 4), with a median duration of intracranial response of 7.2 months [84]. Despite being promising, the performance of T-DXd in active BMs was assessed in very small populations, so studies enrolling larger populations are needed.

3.2.3. Sacituzumab Govitecan

The randomized phase III ASCENT trial investigated the efficacy of SG compared with that of single-agent TPC (eribulin, vinorelbine, capecitabine, or gemcitabine) in patients with relapsed or refractory metastatic triple-negative breast cancer (mTNBC) [85]. In this trial, patients with stable BMs were eligible but not included in the primary endpoint analysis; these patients may constitute a maximum of 15% of the overall trial population. In the subgroup analysis of the 61 patients with BMs at study entry, a trend toward improvement in mPFS was demonstrated in favor of SG vs. TPC (2.8 months vs. 1.6 months; HR 0.68; 95% CI 0.38–1.23), however without any benefits for OS (7.0 vs. 7.5 months, respectively; HR 0.96; 95% CI 0.55–1.68) [86].
Some of the most relevant ongoing trials on ADC in mBC are listed in Table 6. Data on intracranial efficacy are not available as of today.

4. Combination with Radiation Therapy

Given the relatively recent introduction of ADCs in clinical practice, there is a current lack of knowledge about the potential synergy with other anticancer treatment strategies, such as RT.
The concomitant use of ADCs and RT has not been assessed in prospective clinical trials so far. Historically, the rationale for combining chemotherapy and RT stems, among other mechanisms, from the ability of some chemotherapeutic agents to block the cell cycle in the M, late G1 and S phases, in which cells are most susceptible to radiation damage (5-FU, gemcitabine) [87].
Given the mechanism of action of ADCs, it seems reasonable that the same effects could be obtained by the toxic payloads combined with RT. Most of the scarce data available come from BC, in particular from the use of T-DM1, the first ADC approved for breast cancer. No increased toxicity was reported with the concomitant administration of T-DM1 and whole-brain RT (WBRT) in case reports and preclinical cellular models [88,89,90,91].
In contrast, the use of T-DM1 concomitant to stereotactic radiosurgery SRS seems to come with an increased risk of neurological side effects, including clinically significant radionecrosis expansive hematoma in delayed cerebral radiation necrosis [92,93], intracranial hemorrhages [94], and brain edema [95,96].
One possible mechanism underlying this increased toxicity is an off-tumor on-target effect, driven by the expression of HER2 on astrocytes [70,97], which may be targeted by ADCs, with the release of their cytotoxic payloads.
In a large retrospective cohort study conducted by Lebow et al., data on 98 patients with BMs from different cancer types (NSCLC, BC, HER2 amplified Esophageal and/or gastric cancer, HER2 amplified Salivary) who received at least one course of stereotactic RT (SRT) concurrently with T-DM1, T-DXd, or SG were reviewed. SRS was considered concurrent if it was delivered 7 or fewer days before or 21 or fewer days after ADC, and a control group of patients treated with SRS and the ADC sequentially was created for comparison. The concurrent administration of ADC with SRS was associated with a higher risk of symptomatic radionecrosis, both on univariable analysis (SHR, 4.01 [95% CI, 1.79–9.01]; p < 0.001) and multivariable analysis (SHR, 4.31 [95% CI, 1.95–9.50]; p < 0.001), controlling for prior RT (SHR, 2.99 [95% CI, 1.26–7.09]; p = 0.01) and BM volume (SHR, 1.14/cm3 [95% CI, 1.09–1.19]/cm3; p < 0.001). This risk was significantly more pronounced for larger and previously irradiated BMs [98].
Among the concurrent treatment group, the specific ADC was not associated with a risk of radionecrosis (p = 0.74). Given the high rates of local control and its favorable neurocognitive safety profile, SRS remains a key therapeutic option in BMs. However, physicians must consider the increased risk of radionecrosis and monitor patients closely when treating them concurrently with ADCs, or avoid the concomitant use of SRS and ADCs if possible.

5. Discussion

This review aims to summarize the current evidence on ADC therapies in the specific context of the CNS when used against BMs or adult-type gliomas.
Despite their well-recognized lack of systemic therapeutic options, these two populations have historically been underrepresented in clinical trials [99]. The poor prognosis of these patients, coupled with their frequent rapid clinical deterioration and the mechanisms of resistance to most therapeutic strategies, likely explains why patients with symptomatic or active BMs have traditionally been excluded from large, randomized trials. However, their underrepresentation, particularly in early-phase clinical trials, is difficult to justify, as these patients could play a valuable role in assessing the safety and determining the recommended doses of experimental drugs, providing valuable information about the activity of novel compounds at the CNS level [100].
Meanwhile, ADCs have emerged as a new effective class of anticancer agents currently revolutionizing the treatment paradigm of a wide range of different tumor types, attaining seven FDA approvals so far, with numerous trials still underway [101]. Given the very recent advent of ADCs in the anticancer therapeutic armamentarium, most of the data covered in this review are retrospective or derived from post hoc analyses of prospective trials. They are focused on a limited number of cancer histotypes, predominantly NSCLC and BC.
Over the years, the continuous technological evolution of ADCs has seen the development of these new drugs up to their third generation. T-DXd is considered the first exemplary drug of the third-generation anti-HER2 ADCs because of its technological advancements in the linker, DAR, and payload. Its linker allows the conjugation of up to eight cytotoxic payload molecules (DX-d) while not reducing the compound’s half-life [102]. Together with the high membrane permeability of the DX-d compound, it explains the higher intracranial activity of T-DXd compared to that of its second-generation predecessor, T-DM1, in HER2-positive BCBM, with encouraging preliminary results in HER2-low BM tumors [103].
It is more complex to compare the intracranial activity of SG with that of T-DXd in two different subtypes of BC, TNBC and HER2-positive. Both drugs possess the typical characteristics of third-generation ADCs, such as high DAR, cleavable linker, and bystander effect. One possible explanation for the higher T-DXd activity is that it targets an oncogenic driver, whereas SG is directed against a transmembrane protein frequently overexpressed across various carcinomas. However, this contrasts with the promising intracranial results observed in the HERTHENA-Lung01 trial using HER3-DXd in EGFR-mutant NSCLC, where HER3 is unlikely to act as the primary oncogenic driver. This discrepancy highlights the still-unknown mechanisms underlying the variability in intracranial responses to different ADCs.
Intricate is the challenge in gliomas headed by GBM, the most aggressive primary brain malignancy, which has not yet seen any ADC-based therapy approval. We previously underscored the heterogeneity of EGFR amplification and its mutated form EGFRvIII in GBM cells, which could potentially lead to the failure of INTELLANCE-1 trial. Furthermore, Depatux-M requires complete lysosomal degradation to function, as it does not have a cleavable linker, and the MMAF payload hardly crosses intact cell membranes [104]. These technical features may have resulted in the early selection of resistant clones and justified the loss of survival advantage. In contrast to BMs, the diffuse infiltrative nature of glioblastoma cells, especially in the peripheral area where the BBB may retain its physiological functions, could pose a substantial hurdle to ADC efficacy.
One potential strategy is to move away from the traditional mAb structure and couple the payload to a polypeptide or single-chain variable fragment with a smaller molecular weight, enhancing penetration efficiency and payload delivery to tumor tissues [105]. Another possible approach proposed in a study led by Anami et al. involves optimizing ADCs with homogeneous DAR values, which were shown to achieve higher brain bioavailability. High-DAR components in heterogeneous ADCs demonstrate poor CNS penetration, leading to reduced effective DAR compared to that of their homogeneous counterpart [106]. They prove that ADCs with high DAR have greater hydrophobicity, form macromolecules and are characterized by reduced plasma stability, leading to impaired BTB penetration and increased off-target toxicity [107].
Traditional ADCs that utilize cysteine or lysine amino acids as attachment points exhibit significant variability in DAR. In contrast, site-specific conjugation techniques, such as THIOMAB, can lead to the generation of homogeneous ADCs characterized by a constant DAR, by exploiting the insertion of cysteine residues at different positions at the light and heavy chains for conjugation [108]. Several other conjugation techniques are being investigated, including the use of microbial transglutaminase, an enzyme that normally catalyzes post-translational modifications of proteins, which can facilitate the conjugation process and provide flexibility in the selection of the conjugation site and DAR [109]. Another promising technique is AJICAP-M, a traceless affinity peptide-mediated conjugation strategy, which can produce more stable ADCs with minimized payload detachment, suggesting a higher therapeutic index than that of conventional ADCs [110].
For this reason, manufacturing homogeneous ADCs with optimized DAR can potentially increase payload delivery in pBTs and BMs.
The bystander effect also appears to be important in determining the activity of ADCs at the CNS level by allowing cytotoxic activity even against target-negative cells. The choice of a payload that is small, lipophilic and that is not a substrate of the P-gp could be a strategy to allow a better penetration at the CNS level. This could in part explain the higher activity of T-DXd compared to that of T-DM1, the payload of the latter being a substrate of the efflux systems of the BBB [111].
Besides the payload, the type and stability of the linker are of paramount importance.
New techniques including exo-linkers, with their two-step activation system, and tandem linkers incorporating several cleavage steps have the potential to increase plasma stability and reduce premature payload release, potentially reducing systemic toxicities and enabling better payload distribution within the CNS [112,113,114].
Leptomeningeal disease is still an entity excluded from most prospective clinical trials, and current systemic treatment approaches are derived from BMs. In cohort 5 of the DEBBRAH trial, T-DXd was tested on a total of 7 patients with untreated leptomeningeal involvement, leading to an mOS of 13.3 months and an mPFS of 11.9 months. Of the five patients who progressed, none of them had intracranial progression or worsening of symptoms referable to leptomeningeal carcinomatosis. Although these data are from a very small number of patients and should be confirmed by further larger prospective studies, they provide an early indication of the efficacy of ADCs in the treatment of leptomeningeal carcinomatosis [115].
At the methodological level, the prospective TUXEDO-1 and DEBBRAH trials currently constitute the most rigorous and statistically appropriate frameworks for the dedicated evaluation of intracranial activity-associated ADCs. In TUXEDO-1, baseline patient characteristics are comprehensively delineated, with particular attention to neurological symptomatology, prior local therapeutic interventions, and the dynamic status of intracranial disease, distinguishing between progressive and newly diagnosed lesions. The DEBBRAH trial adopts an even more granular stratification strategy, enrolling patients into five discrete cohorts according to HER2 membrane receptor expression, previous intracranial treatment history, the presence or absence of neurological symptoms, and leptomeningeal dissemination. Such meticulous cohorting facilitates a highly refined interrogation of ADC efficacy within clinically heterogeneous populations, addressing variables often overlooked in broader trial designs. The remaining studies have some critical issues as they were not formally designed to study the intracranial activity of ADCs. While approximately 75% of studies report including CNS-specific endpoints, only a minority, one-third, permit the enrollment of patients with active or progressing intracranial disease.
Another key issue highlighted by the studies collected in this review and limiting the ability to analyze data is the high heterogeneity of criteria used for assessing intracranial response in BMs. Slightly more than 25% of the studies reported using the RANO-BM criteria [99], which should represent the gold standard. The remainder predominantly saw the use of RECIST 1.1 or RECIST 1.0 criteria or the investigator’s choice. It would be ideal to report stratified data for patients with stable and active BMs, as this would improve result clarity and refine clinical indications. This stratification is essential for distinguishing the causal impact of intracranial responses due to systemic therapy, prior local therapy, or the interplay between both treatment modalities. Additionally, a key factor in interpreting intracranial response data is the standardization of eligibility criteria, particularly regarding the washout period from the last local therapy in cases of pretreated BMs.
The data gathered in this review demonstrate remarkable consistency for HER2- + BCBM and provide valuable guidance for clinical decision-making. Whether pretreated or not, the outstanding efficacy of T-DXd in stable HER-2 + BMs suggests it may be preferred over other active regimens such as T-DM1 or the Trastuzumab–Tucatinib–Capecitabine combination [74,78,116]. However, despite encouraging findings from the Destiny breast-12, DEBBRAH, and TUXEDO trials, the evidence remains too premature to recommend T-DXd for patients with active or progressing brain metastases. Ideally, a randomized trial would be the gold standard with which to determine whether systemic therapy with ADCs alone in BMs can achieve superior outcomes, potentially delaying or even reducing the need for local interventions.

6. Conclusions

ADCs are ready to become a key component of clinical practice for BC and NSCLC with active or stable brain metastases, offering the potential to delay other systemic or local therapies while improving survival outcomes. In pBTs, the landscape for ADCs remains more complex due to the unique biological challenges of these diseases. However, advancements in next-generation ADCs are likely to overcome these obstacles, paving the way for more effective treatment strategies in both primary and metastatic brain tumors.

Author Contributions

Conceptualization, methodology, and writing—original draft preparation, F.B., C.B. and A.D.M.; data curation and writing—review and editing, A.D., P.P., A.L. and I.T.; data curation, A.D; supervision, F.P., P.N. and A.S.; writing—review and editing and supervision, M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

Appendix A

Table A1. Summary of ongoing clinical trials evaluating ADCs in NSCLC patients.
Table A1. Summary of ongoing clinical trials evaluating ADCs in NSCLC patients.
Clinical TrialResponse EndpointADCTargetLinker TypePayload ActionPopulationCNS Metastasis
Eligibility
Trial TypeStatus
HER3-DXd in Breast Cancer and NSCLC Brain Metastases and Solid Tumor Leptomeningeal Disease (TUXEDO-3) (NCT05865990)iORR as defined by RANO -BM criteriaPatritumab deruxtecan (HER3-DXd)HER3CleavableTopoisomerase
I inhibitor
(Deruxtecan)
Patients with previously treated a/m NSCLC with
newly diagnosed or progressive BM
Patients are eligible if steroid use is <8 mg of Dexamethasona/dayPhase IIActive, not recruiting
Datopotamab Deruxtecan (Dato-DXd) for Non-Small Cell Lung Cancer (NSCLC) patients with Active Brain Metastases (TUXEDO-5) (NCT06676917)iORR as defined by RANO-BM criteriaDatopotamab Deruxtecan (Dato-DXd)TROP2CleavableTopoisomerase
I inhibitor
(Deruxtecan)
Patients with previously treated a/m non squamous NSCLC with active BMPatients with BM and leptomeningeal disease are allowed if there is no indication for immediate local therapyPhase IIRecruitment not yet open; the estimated study start date is April 2025
Sacituzumab Tirumotecan (MK-2870) Versus Chemotherapy in Previously Treated a/m Nonsquamous NSCLC with EGFR Mutations or Other Genomic Alterations (NCT06074588)PFS as defined by RECIST v 1.1 and
OS
Sacituzumab tirumotecan
(MK-2870)
TROP2CleavableTopoisomerase I
Inhibitor
(Tirumotecan)
Patients with a/m non-squamous NSCLC
with treatment-refractory
progressive disease
Previously treated
BM included; active
BM and
leptomeningeal disease excluded
Phase IIIRecruiting
Phase III, Open-label, First-line Study of Dato-DXd in Combination with Durvalumab and Carboplatin for Advanced NSCLC Without Actionable Genomic Alterations (AVANZAR) (NCT05687266)PFS as defined by RECIST v1.1 and OSDatopotamab Deruxtecan (Dato-DXd)TROP2ClevableTopoisomerase
I inhibitor
(Deruxtecan)
Patients with a/m NSCLC without actionable
genomic alterations with approved and a viable target therapy
Active BM and leptomeningeal disease excludedPhase IIIActive, not recruiting
Study of Pembrolizumab (MK-3475) Monotherapy Versus Sacituzumab Govitecan in Combination with Pembrolizumab for Participants With Metastatic Non-small Cell Lung Cancer (NSCLC) With Programmed Cell Death Ligand 1 (PD-L1) Tumor Proportion Score (TPS) ≥ 50% (MK-3475-D46) (NCT05609968)PFS as defined by RECIST v1.1 and OSSacituzumab govitecan (SG)TROP2CleavableTopoisomerase I
Inhibitor (govitecan)
Patients with a/m NSCLC without an indication of EGFR-ALK-1, or ROS-1-targeted therapies with PD-L1 expression > 50%Active BM and leptomeningeal disease excludedPhase IIIRecruiting
Study of Dato-DXd Plus Pembrolizumab vs. Pembrolizumab Alone in the First-line Treatment of Subjects with Advanced or Metastatic NSCLC Without Actionable Genomic Alterations (TROPION-Lung08)
(NCT05215340)
PFS as defined by RECIST v1.1 and OSDatopotamab
Deruxtecan (Dato-DXd)
TROP2CleavableTopoisomerase I
Inhibitor
(Deruxtecan)
Patients with a/m non-squamous NSCLC without actionable genomic alterations (mKRAS eligible) with PD-L1 expression > 50%Active and untreated BM excluded; leptomeningeal disease excludedPhase IIIRecruiting
A Study of YL202 in Selected Patients with Advanced Solid Tumors
(NCT06107686)
ORR as defined by RECIST v1.1YL202HER3CleavableTopoisomerase I
Inhibitor
(YL0010014)
Patients with a/m NSCLCPatients with BM excludedPhase IIRecruiting
A Study of Disitamab
Vedotin in Previously
Treated Solid Tumors That
Express HER2
(NCT06003231)
ORR as defined by RECIST v1.1Disitamab
Vedotin
HER2CleavableAuristatin
microtubule
inhibitor
(monomethyl auristatin E)
Patients with a/m solid tumors, including
NSCLC, with HER2 overexpression
Active and
untreated BM and leptomeningeal disease excluded
Phase IIRecruiting
A Phase 2 Study of BA3011 Alone and in Combination with PD-1 Inhibitor in Adult Patients with Metastatic Non-small Cell Lung Cancer (NSCLC) Who Had Prior Disease Progression on a PD-1/L-1 Inhibitor, EGFR, or ALK Inhibitor.
(NCT04681131)
ORR as defined by RECIST v1.1CAB-AXL-ADCAXLCleavableAuristatin
microtubule
inhibitor
(monomethyl auristatin E)
Patients with a/m NSCLCPatients with active BM excludedPhase IIActive, non recruiting
Datopotamab Deruxtecan (Dato-DXd, DS-1062a) in Advanced and/or Unresectable Non-Small Cell Lung Cancer (ICARUS-LUNG01) (NCT04940325)ORR as defined by RECIST v1.1Datopotomab Deruxtecan
(Dato-DXd)
TROP2CleavableTopoisomerase I
Inhibitor
(Deruxtecan)
Patients with a/m NSCLCBM eligible only if treated and clinically stable without medical treatmentPhase IIActive, non recruiting
CAB-ROR2-ADC Safety and
Efficacy Study in Patients
with TNBC or Head & Neck
Cancer (Ph1) and NSCLC or
Melanoma (Ph2) (NCT03504488)
ORR Ozuriftamab
vedotin
ROR2CleavableAuristatin
microtubule
inhibitor
(monomethyl auristatin E)
Patients with a/m solid tumors, including NSCLC, that is refractory or not amenable to the standard-of-care therapyUncontrolled BM
excluded
Phase I/IIActive, non recruiting
A Clinical Trial of TQB2102 for Injection in Non-small Cell Lung Cancer with HER2 Gene Abnormality (NCT06496490)ORRTQB2102HER2CleavableTopoisomerase I
Inhibitor
(topotecan)
Patients with a/m NSCLC refractory
to standard-of-care therapy
Stable BM included
Uncontrolled BM or Leptomeningeal disease excluded
Phase IIRecruiting
Study to Investigate Luveltamab Tazevibulin in Adults with Advanced or Metastatic Non-small Cell Lung Cancer (NCT06555263)ORR as defined by RECIST v1.1Luveltamab TazevibulinFOLR1CleavableAuristatin
microtubule
inhibitor (hemiasterina)
Patients who are a/m NSCLC-positive per FOLR1 expression per central testingUntreated BM are excludedPhase IIRecruiting
A First-in-human Study Using BDC-1001 as a Single Agent and in Combination with Nivolumab in Advanced HER2-Expressing Solid Tumors (NCT04278144)ORRBDC-1001HER2Non cleavableISAC (Trastuzumab conjugated with TLR7/8 agonist)Patients with an a/m solid tumor with documented HER2-protein expression or gene amplification for which approved therapies have been exhausted or not clinically indicatedUntreated BM or active BM are excludedPhase I/IIActive, non recruiting
Phase I–II, FIH, TROP2 ADC, Advanced Unresectable/Metastatic Solid Tumors, Refractory to Standard Therapies (A264) (NCT04152499)ORR (phase II)SKB264TROP2CleavableMicrotubule
inhibitor
(Belotecan-based payload)
Patients with a/m solid tumors, including NSCLC, that is refractory or not amenable to the standard-of care-therapySymptomatic BM or any radiation or surgery for BMs within 1 month of first infusion of study drug are the exclusion criteria (Phase I). Patients with active BM or leptomeningeal disease are excluded (Phase II)Phase I/IIRecruiting
PRO1184 for Advanced Solid
Tumors (PRO1184-001) (NCT05579366)
ORR as defined by Recist v1.1 (phase II)PRO1184FOLR1CleavableTopoisomerase I
Inhibitor (exatecan)
Patients with a/m solid tumors, including NSCLC, that is refractory or not amenable to the standard-of-care therapyActive BMs are excludedPhase I/IIRecruiting
Study of DF1001 in Patients with Advanced Solid Tumors (NCT04143711)ORR as defined by RECIST v1.1DF1001HER2CleavableInduce cell death by targeting NK cells and T-cell activation signalsA/m NSCLC patients with HER2 overexpression/amplification/mutation who progressed after a platinum-based CT and have received and progressed on or after anti PD-(L)1 therapy.
Those for whom a/m NSCLC progressed during or after platinum doublet-based chemotherapy (expansion NSCLC cohort)
Active BMs or a history of BM are the exclusion criteriaPhase I/IIRecruiting
Study of REGN5093-M114
(METxMET Antibody–Drug
Conjugate) in Adult Patients
with Mesenchymal–Epithelial
Transition Factor (MET)
Overexpressing Advanced
Cancer
(NCT04982224)
ORR
(Phase II)
REGN5093-M114METxMETCleavableAuristatin
microtubule
inhibitor (M24)
Patients with a/m NSCLC with MET
Overexpression
Untreated/active BM and
leptomeningeal disease excluded
Phase I/IIRecruiting
Clinical Study of
Antibody–Drug Conjugate
MYTX-011 in Subjects with
Non-Small Cell Lung Cancer
(NCT05652868)
ORR, OS and PFS are secondary endpointsMYTX-011METClevableAuristatin
microtubule
inhibitor (Monomethyl auristatin E)
Patients with a/m NSCLC with MET alterations that is refractory to the standard-of-care therapyUntreated/active BM and leptomeningeal disease excludedPhase IRecruiting
An Efficacy and Safety Study
of Cofetuzumab Pelidotin in
Participants with
PTK7-Expressing, Recurrent
Non-Small Cell Lung Cancer (NCT04189614)
OS, PFS and DOR are secondary endpointsCofetuzumab
pelidotin
PTK7CleavableAuristatin
microtubule
inhibitor (Auristatin-0101)
Patients with recurrent and treatment-refractory
NSCLC with PTK7 expression
Asymptomatic and
treated BM
included, active
BM excluded
Phase IActive, non recruiting
A Study to Evaluate
TROP2 ADC LCB84 Single
Agent and in Combination
with an Anti-PD-1 Ab in
Advanced Solid Tumors (NCT05941507)
PFS, ORR, DOR, TTP as defined by RECIST v1.1, iRECIST, and RANO-BM
OS
LCB84TROP2CleavableMicrotubule
inhibitor (Monomethyl auristatin E)
Patients with advanced solid tumors, including NSCLC that is refractory or not amenable to the standard-of-care therapyStable and treated BM includedPhase I/IIRecruiting
DS8201a and
Pembrolizumab in
Participants with Locally
Advanced/Metastatic Breast
or Non-Small Cell Lung
Cancer
(NCT04042701)
ORRTrastuzumab
deruxtecan
HER2CleavableTopoisomerase I
Inhibitor
(Deruxtecan)
Patients with a/m breast cancer or NSCLC with HER2 overexpression or HER2-mutant diseaseActive BM excludedPhase IRecruiting
Phase Ib Study of the Safety
of T-DXd and
Immunotherapy Agents with
and Without Chemotherapy
in Advanced or Metastatic
HER2-positive, non-squamous
NSCLC (DL03)
(NCT04686305)
PFS, ORR, DOR, as defined by RECIST v1.1,
OS
Trastuzumab
Deruxtecan (T-DXd)
HER2CleavableTopoisomerase I
Inhibitor
(Deruxtecan)
Patients with a/m non-squamous NSCLC that are refractory to systemic therapy or
treatment-naive with HER2 overexpression
Untreated and
symptomatic BM
excluded
Phase IRecruiting
A Study of LY4052031 in
Participants with Advanced
or Metastatic Urothelial
Cancer or Other Solid
Tumors (NEXUS-01) (NCT06465069)
ORR defined by RECIST v1.1LY4052031NECTIN4CleavableTopoisomerase I
Inhibitor
(Camptothecin-98)
Patients with a/m solid tumors, including NSCLC that is refractory or not amenable
to the standard-of-care therapy
Known or suspected
uncontrolled BM excluded
Phase IRecruiting
A Study of LY4170156 in
Participants with Selected
Advanced Solid Tumors (NCT06400472)
ORR defined by RECIST v1.1LY4170156FOLR1CleavableTopoisomerase I
Inhibitor
(Camptothecin-98)
Patients with advanced solid tumors, including NSCLC that is refractory or not amenable to the standard-of-care
therapy
Active and
untreated BM, and
history of leptomeningeal disease
excluded
Phase IRecruiting
A Study of LY4101174 in
Participants with Recurrent,
Advanced or Metastatic
Solid Tumors
(NCT06238479)
ORR defined by RECIST v1.1 (primary endpoint)
DOR, PFS, TTR, DCF as defined by RECIST v1.1
OS
LY4101174NECTIN4CleavableTopoisomerase I
Inhibitor
(Camptothecin-98)
Patients with a/m solid tumors, including NSCLC that is refractory or not amenable
to the standard-of-care therapy
Known or suspected
uncontrolled BM excluded
Phase IRecruiting
Safety of GQ1001 in Adult
Patients with HER2-Positive
Advanced Solid Tumors (NCT04450732)
ORR, DCR, PFS and DoR as defined by RECIST v1.1GQ1001HER2CleavableMicrotubule
inhibitor (Maytansinoide DM1)
Patients with a/m HER2-expressing solid tumors, including NSCLC that is refractory or not amenable to the standard-of-care therapyTreated and
asymptomatic BM
included untreated
and symptomatic
BM excluded
Phase IRecruiting
Phase I–II, FIH, TROP2 ADC, Advanced
Unresectable/Metastatic
Solid Tumors, Refractory to Standard Therapies (A264) (NCT04152499)
ORR, DoR, PFS as defined by RECIST v1.1
OS
Sacituzumab
tirumotecan
TROP2CleavableTopoisomerase I inhibitor (belotecan derivative)Patients with a/m solid tumors, including metastatic NSCLC, that is refractory or not amenable to the standard of-care-therapySymptomatic and
active BM, history of
LMD, brainstem
metastasis, and
spinal cord
metastasis excluded
Phase I/IIRecruiting
Phase 1 Study of
CPO301 in Adult Patients
with Advanced or Metastatic
Solid Tumors (NCT05948865)
Efficacy assesment not specifiedCPO301EGFRCleavableNon-specifiedPatients with a/m solid tumors, including
metastatic NSCLC, that is
refractory or not amenable
to the standard-of-care
therapy
Known, active, or uncontrolled BM or leptomeningeal disease excludedPhase IRecruiting
A Study of PF-08046050
(SGN-CEACAM5C) in
Adults with Advanced Solid
Tumors
(NCT06131840)
ORR, DoR, PFS as defined by RECIST v1.1
Best response
OS
PF-08046050CEACAM5CleavableTopoisomerase I
inhibitor
Patients with a/m solid tumors, including
metastatic NSCLC, that is
refractory or not amenable
to the standard-of-care
therapy
Stable and treated
BM included
Phase IRecruiting
PRO1184 for Advanced Solid
Tumors (PRO1184-001) (NCT05579366)
ORR, DoR, PFS as defined by RECIST v1.1
OS
PRO1184FOLR1CleavableTopoisomerase I
inhibitor
Patients with a/m solid tumors, including
NSCLC that is refractory
or not amenable to the
standard-of-care therapy
Previously treated
and stable BM
included active
BM or leptomeningeal diseaseexcluded
Phase I/IIRecruiting
Abbreviations: ADC, antibody–drug conjugate; a/m, advanced/metastatic; AXL, AXL receptor tyrosine kinase; BM, brain metastasis; CEACAM 5, Carcinoembryonic Antigen-Related Cell Adhesion Molecule 5; DCR, disease control rate; DoR, duration of response; Dxd, Deruxtecan; EGFR, epidermal growth factor receptor; FOLR1, Folate Receptor Alpha; HER2, human epidermal growth factor receptor 2; HER3, human epidermal growth factor receptor 3; iDCR, intracranial disease control rate; iORR, intracranial overall response rate; MET, mesenchymal–epithelial transition factor; Nectin 4, Nectin Cell Adhesion Molecule 4; NSCLC, non-small cell lung cancer; OS, overall survival; PFS, progression-free survival; PTK7, Protein Tyrosine Kinase 7; ROR2, receptor tyrosine kinase-like orphan receptor 2; TF, tissue factor; TROP2, trophoblast cell-surface antigen 2.

References

  1. Brown, N.F.; Ottaviani, D.; Tazare, J.; Gregson, J.; Kitchen, N.; Brandner, S.; Fersht, N.; Mulholland, P. Survival Outcomes and Prognostic Factors in Glioblastoma. Cancers 2022, 14, 3161. [Google Scholar] [CrossRef] [PubMed]
  2. Sperduto, P.W.; Mesko, S.; Li, J.; Cagney, D.; Aizer, A.; Lin, N.U.; Nesbit, E.; Kruser, T.J.; Chan, J.; Braunstein, S.; et al. Survival in Patients with Brain Metastases: Summary Report on the Updated Diagnosis-Specific Graded Prognostic Assessment and Definition of the Eligibility Quotient. J. Clin. Oncol. 2020, 38, 3773–3784. [Google Scholar] [CrossRef] [PubMed]
  3. Ostrom, Q.T.; Price, M.; Neff, C.; Cioffi, G.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2015–2019. Neuro-Oncol. 2022, 24, v1–v95. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, L.; Ma, J.; Zou, Z.; Liu, H.; Liu, C.; Gong, S.; Gao, X.; Liang, G. Clinical Characteristics and Prognosis of Patients with Glioblastoma: A Review of Survival Analysis of 1674 Patients Based on SEER Database. Medicine 2022, 101, e32042. [Google Scholar] [CrossRef]
  5. Bondy, M.L.; Scheurer, M.E.; Malmer, B.; Barnholtz-Sloan, J.S.; Davis, F.G.; Il’yasova, D.; Kruchko, C.; McCarthy, B.J.; Rajaraman, P.; Schwartzbaum, J.A.; et al. Brain Tumor Epidemiology: Consensus from the Brain Tumor Epidemiology Consortium (BTEC). Cancer 2008, 113, 1953–1968. [Google Scholar] [CrossRef]
  6. Achrol, A.S.; Rennert, R.C.; Anders, C.; Soffietti, R.; Ahluwalia, M.S.; Nayak, L.; Peters, S.; Arvold, N.D.; Harsh, G.R.; Steeg, P.S.; et al. Brain Metastases. Nat. Rev. Dis. Primers 2019, 5, 5. [Google Scholar] [CrossRef] [PubMed]
  7. Dipasquale, A.; Barigazzi, C.; Losurdo, A.; Persico, P.; Di Muzio, A.; Navarria, P.; Pessina, F.; van den Bent, M.; Santoro, A.; Simonelli, M. Brain Metastases and Next-Generation Anticancer Therapies: A Survival Guide for Clinicians. Crit. Rev. Oncol. Hematol. 2024, 194, 104239. [Google Scholar] [CrossRef]
  8. Alemany, M.; Velasco, R.; Simó, M.; Bruna, J. Late Effects of Cancer Treatment: Consequences for Long-Term Brain Cancer Survivors. Neuro-Oncol. Pract. 2020, 8, 18–30. [Google Scholar] [CrossRef]
  9. Makale, M.T.; McDonald, C.R.; Hattangadi-Gluth, J.A.; Kesari, S. Mechanisms of Radiotherapy-Associated Cognitive Disability in Patients with Brain Tumours. Nat. Rev. Neurol. 2017, 13, 52–64. [Google Scholar] [CrossRef]
  10. Brenner, A.W.; Patel, A.J. Review of Current Principles of the Diagnosis and Management of Brain Metastases. Front. Oncol. 2022, 12, 857622. [Google Scholar] [CrossRef]
  11. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The Blood-Brain Barrier and Blood-Tumour Barrier in Brain Tumours and Metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef]
  12. Freskgård, P.-O.; Urich, E. Antibody Therapies in CNS Diseases. Neuropharmacology 2017, 120, 38–55. [Google Scholar] [CrossRef] [PubMed]
  13. Marcucci, F.; Corti, A.; Ferreri, A.J.M. Breaching the Blood–Brain Tumor Barrier for Tumor Therapy. Cancers 2021, 13, 2391. [Google Scholar] [CrossRef]
  14. Chhichholiya, Y.; Ruthuparna, M.; Velagaleti, H.; Munshi, A. Brain Metastasis in Breast Cancer: Focus on Genes and Signaling Pathways Involved, Blood-Brain Barrier and Treatment Strategies. Clin. Transl. Oncol. 2023, 25, 1218–1241. [Google Scholar] [CrossRef] [PubMed]
  15. ESMO. Trastuzumab Deruxtecan Shows High Intracranial Response Rate in Patients with Active Brain Metastases from HER2-Positive Breast Cancer. Available online: https://www.esmo.org/oncology-news/trastuzumab-deruxtecan-shows-high-intracranial-response-rate-in-patients-with-active-brain-metastases-from-her2-positive-breast-cancer (accessed on 20 March 2024).
  16. Fu, Z.; Li, S.; Han, S.; Shi, C.; Zhang, Y. Antibody Drug Conjugate: The “Biological Missile” for Targeted Cancer Therapy. Signal Transduct. Target. Ther. 2022, 7, 93. [Google Scholar] [CrossRef] [PubMed]
  17. Staudacher, A.H.; Brown, M.P. Antibody Drug Conjugates and Bystander Killing: Is Antigen-Dependent Internalisation Required? Br. J. Cancer 2017, 117, 1736–1742. [Google Scholar] [CrossRef]
  18. Mckertish, C.; Kayser, V. Advances and Limitations of Antibody Drug Conjugates for Cancer. Biomedicines 2021, 9, 872. [Google Scholar] [CrossRef]
  19. Cortés, J.; Hurvitz, S.A.; Im, S.-A.; Iwata, H.; Curigliano, G.; Kim, S.-B.; Chiu, J.W.Y.; Pedrini, J.L.; Li, W.; Yonemori, K.; et al. Trastuzumab Deruxtecan versus Trastuzumab Emtansine in HER2-Positive Metastatic Breast Cancer: Long-Term Survival Analysis of the DESTINY-Breast03 Trial. Nat. Med. 2024, 30, 2208–2215. [Google Scholar] [CrossRef]
  20. Marin, B.-M.; Porath, K.A.; Jain, S.; Kim, M.; Conage-Pough, J.E.; Oh, J.-H.; Miller, C.L.; Talele, S.; Kitange, G.J.; Tian, S.; et al. Heterogeneous Delivery across the Blood-Brain Barrier Limits the Efficacy of an EGFR-Targeting Antibody Drug Conjugate in Glioblastoma. Neuro-Oncol. 2021, 23, 2042–2053. [Google Scholar] [CrossRef]
  21. Siegall, C.B. Targeted Toxins as Anticancer Agents. Cancer 1994, 74, 1006–1012. [Google Scholar] [CrossRef]
  22. Li, Y.M.; Hall, W.A. Targeted Toxins in Brain Tumor Therapy. Toxins 2010, 2, 2645–2662. [Google Scholar] [CrossRef]
  23. Sampson, J.H.; Akabani, G.; Archer, G.E.; Berger, M.S.; Coleman, R.E.; Friedman, A.H.; Friedman, H.S.; Greer, K.; Herndon, J.E.; Kunwar, S.; et al. Intracerebral Infusion of an EGFR-Targeted Toxin in Recurrent Malignant Brain Tumors. Neuro-Oncol. 2008, 10, 320–329. [Google Scholar] [CrossRef]
  24. Zhu, H.; Acquaviva, J.; Ramachandran, P.; Boskovitz, A.; Woolfenden, S.; Pfannl, R.; Bronson, R.T.; Chen, J.W.; Weissleder, R.; Housman, D.E.; et al. Oncogenic EGFR Signaling Cooperates with Loss of Tumor Suppressor Gene Functions in Gliomagenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 2712–2716. [Google Scholar] [CrossRef] [PubMed]
  25. Lassman, A.B.; Aldape, K.D.; Ansell, P.J.; Bain, E.; Curran, W.J.; Eoli, M.; French, P.J.; Kinoshita, M.; Looman, J.; Mehta, M.; et al. Epidermal Growth Factor Receptor (EGFR) Amplification Rates Observed in Screening Patients for Randomized Trials in Glioblastoma. J. Neuro-Oncol. 2019, 144, 205–210. [Google Scholar] [CrossRef] [PubMed]
  26. Narita, Y.; Muragaki, Y.; Kagawa, N.; Asai, K.; Nagane, M.; Matsuda, M.; Ueki, K.; Kuroda, J.; Date, I.; Kobayashi, H.; et al. Safety and Efficacy of Depatuxizumab Mafodotin in Japanese Patients with Malignant Glioma: A Nonrandomized, Phase 1/2 Trial. Cancer Sci. 2021, 112, 5020–5033. [Google Scholar] [CrossRef] [PubMed]
  27. An, Z.; Aksoy, O.; Zheng, T.; Fan, Q.-W.; Weiss, W.A. Epidermal Growth Factor Receptor and EGFRvIII in Glioblastoma: Signaling Pathways and Targeted Therapies. Oncogene 2018, 37, 1561–1575. [Google Scholar] [CrossRef]
  28. Kunwar, S.; Chang, S.; Westphal, M.; Vogelbaum, M.; Sampson, J.; Barnett, G.; Shaffrey, M.; Ram, Z.; Piepmeier, J.; Prados, M.; et al. Phase III Randomized Trial of CED of IL13-PE38QQR vs. Gliadel Wafers for Recurrent Glioblastoma. Neuro-Oncol. 2010, 12, 871–881. Available online: https://academic.oup.com/neuro-oncology/article/12/8/871/1073747 (accessed on 29 August 2024). [CrossRef]
  29. Sampson, J.H.; Singh Achrol, A.; Aghi, M.K.; Bankiewicz, K.; Bexon, M.; Brem, S.; Brenner, A.; Chandhasin, C.; Chowdhary, S.; Coello, M.; et al. Targeting the IL4 Receptor with MDNA55 in Patients with Recurrent Glioblastoma: Results of a Phase IIb Trial. Neuro-Oncol. 2023, 25, 1085–1097. [Google Scholar] [CrossRef]
  30. Medicenna Therapeutics Corp. Medicenna Announces Compelling Survival Benefit from Phase 2b Study of Bizaxofusp in Recurrent Glioblastoma at the 28th Annual Meeting of the Society for NeuroOncology; Medicenna Therapeutics Corp: Toronto, ON, Canada, 2023; Available online: https://www.globenewswire.com/news-release/2023/11/17/2782496/0/en/Medicenna-Announces-Compelling-Survival-Benefit-from-Phase-2b-Study-of-Bizaxofusp-in-Recurrent-Glioblastoma-at-the-28th-Annual-Meeting-of-the-Society-for-NeuroOncology.html (accessed on 1 February 2025).
  31. Tortorella, S.; Karagiannis, T.C. Transferrin Receptor-Mediated Endocytosis: A Useful Target for Cancer Therapy. J. Membr. Biol. 2014, 247, 291–307. [Google Scholar] [CrossRef]
  32. Gan, H.K.; Burgess, A.W.; Clayton, A.H.A.; Scott, A.M. Targeting of a Conformationally Exposed, Tumor-Specific Epitope of EGFR as a Strategy for Cancer Therapy. Cancer Res. 2012, 72, 2924–2930. [Google Scholar] [CrossRef]
  33. van den Bent, M.; Gan, H.K.; Lassman, A.B.; Kumthekar, P.; Merrell, R.; Butowski, N.; Lwin, Z.; Mikkelsen, T.; Nabors, L.B.; Papadopoulos, K.P.; et al. Efficacy of Depatuxizumab Mafodotin (ABT-414) Monotherapy in Patients with EGFR-Amplified, Recurrent Glioblastoma: Results from a Multi-Center, International Study. Cancer Chemother. Pharmacol. 2017, 80, 1209–1217. [Google Scholar] [CrossRef] [PubMed]
  34. Reardon, D.A.; Lassman, A.B.; Van Den Bent, M.; Kumthekar, P.; Merrell, R.; Scott, A.M.; Fichtel, L.; Sulman, E.P.; Gomez, E.; Fischer, J.; et al. Efficacy and Safety Results of ABT-414 in Combination with Radiation and Temozolomide in Newly Diagnosed Glioblastoma. Neuro-Oncol. 2017, 19, 965–975. Available online: https://academic.oup.com/neuro-oncology/article/19/7/965/2760198 (accessed on 29 August 2024). [CrossRef] [PubMed]
  35. Gan, H.K.; Reardon, D.A.; Lassman, A.B.; Merrell, R.; van den Bent, M.; Butowski, N.; Lwin, Z.; Wheeler, H.; Fichtel, L.; Scott, A.M.; et al. Safety, Pharmacokinetics, and Antitumor Response of Depatuxizumab Mafodotin as Monotherapy or in Combination with Temozolomide in Patients with Glioblastoma. Neuro-Oncol. 2018, 20, 838–847. [Google Scholar] [CrossRef]
  36. Lassman, A.B.; van den Bent, M.J.; Gan, H.K.; Reardon, D.A.; Kumthekar, P.; Butowski, N.; Lwin, Z.; Mikkelsen, T.; Nabors, L.B.; Papadopoulos, K.P.; et al. Safety and Efficacy of Depatuxizumab Mafodotin + Temozolomide in Patients with EGFR-Amplified, Recurrent Glioblastoma: Results from an International Phase I Multicenter Trial. Neuro-Oncol. 2019, 21, 106–114. [Google Scholar] [CrossRef]
  37. Van Den Bent, M.; Eoli, M.; Sepulveda, J.M.; Smits, M.; Walenkamp, A.; Frenel, J.-S.; Franceschi, E.; Clement, P.M.; Chinot, O.; De Vos, F.; et al. INTELLANCE 2/EORTC 1410 Randomized Phase II Study of Depatux-M Alone and with Temozolomide vs. Temozolomide or Lomustine in Recurrent EGFR Amplified Glioblastoma. Neuro-Oncol. 2020, 22, 684–693. [Google Scholar] [CrossRef]
  38. Hoogstrate, Y.; Ghisai, S.A.; de Wit, M.; de Heer, I.; Draaisma, K.; van Riet, J.; van de Werken, H.J.G.; Bours, V.; Buter, J.; Vanden Bempt, I.; et al. The EGFRvIII Transcriptome in Glioblastoma: A Meta-Omics Analysis. Neuro-Oncol. 2022, 24, 429–441. [Google Scholar] [CrossRef] [PubMed]
  39. Lassman, A.B.; Pugh, S.L.; Wang, T.J.C.; Aldape, K.; Gan, H.K.; Preusser, M.; Vogelbaum, M.A.; Sulman, E.P.; Won, M.; Zhang, P.; et al. Depatuxizumab Mafodotin in EGFR-Amplified Newly Diagnosed Glioblastoma: A Phase III Randomized Clinical Trial. Neuro-Oncol. 2023, 25, 339–350. [Google Scholar] [CrossRef]
  40. Padovan, M.; Eoli, M.; Pellerino, A.; Rizzato, S.; Caserta, C.; Simonelli, M.; Michiara, M.; Caccese, M.; Anghileri, E.; Cerretti, G.; et al. Depatuxizumab Mafodotin (Depatux-M) Plus Temozolomide in Recurrent Glioblastoma Patients: Real-World Experience from a Multicenter Study of Italian Association of Neuro-Oncology (AINO). Cancers 2021, 13, 2773. [Google Scholar] [CrossRef]
  41. Hou, J.; Lv, A.; Deng, Q.; Zhang, G.; Hu, X.; Cui, H. TROP2 Promotes the Proliferation and Metastasis of Glioblastoma Cells by Activating the JAK2/STAT3 Signaling Pathway. Oncol. Rep. 2019, 41, 753–764. [Google Scholar] [CrossRef]
  42. Alberti, S.; Trerotola, M.; Moschella, A.; Guerra, E. TROP2 Gene Copy Number Variations Are a Hallmark of Glioblastoma and MEN-2A Cancers. Cancer Res. 2023, 83 (Suppl. S7), 2589. Available online: https://www.researchgate.net/publication/369810215_Abstract_2589_TROP2_gene_copy_number_variations_are_a_hallmark_of_glioblastoma_and_MEN-2A_cancers (accessed on 1 September 2024). [CrossRef]
  43. Guerra, E.; Di Pietro, R.; Stati, G.; Alberti, S. A Non-Mutated TROP2 Fingerprint in Cancer Genetics. Front. Oncol. 2023, 13, 1151090. Available online: https://pubmed.ncbi.nlm.nih.gov/37456256/ (accessed on 1 September 2024). [CrossRef] [PubMed]
  44. Stepan, L.P.; Trueblood, E.S.; Hale, K.; Babcook, J.; Borges, L.; Sutherland, C.L. Expression of Trop2 Cell Surface Glycoprotein in Normal and Tumor Tissues: Potential Implications as a Cancer Therapeutic Target. J. Histochem. Cytochem. 2011, 59, 701–710. [Google Scholar] [CrossRef]
  45. Page, S.; Milner-Watts, C.; Perna, M.; Janzic, U.; Vidal, N.; Kaudeer, N.; Ahmed, M.; McDonald, F.; Locke, I.; Minchom, A.; et al. Systemic Treatment of Brain Metastases in Non-Small Cell Lung Cancer. Eur. J. Cancer 2020, 132, 187–198. [Google Scholar] [CrossRef] [PubMed]
  46. Gillespie, C.S.; Mustafa, M.A.; Richardson, G.E.; Alam, A.M.; Lee, K.S.; Hughes, D.M.; Escriu, C.; Zakaria, R. Genomic Alterations and the Incidence of Brain Metastases in Advanced and Metastatic NSCLC: A Systematic Review and Meta-Analysis. J. Thorac. Oncol. 2023, 18, 1703–1713. [Google Scholar] [CrossRef] [PubMed]
  47. De Carlo, E.; Bertoli, E.; Del Conte, A.; Stanzione, B.; Berto, E.; Revelant, A.; Spina, M.; Bearz, A. Brain Metastases Management in Oncogene-Addicted Non-Small Cell Lung Cancer in the Targeted Therapies Era. Int. J. Mol. Sci. 2022, 23, 6477. [Google Scholar] [CrossRef]
  48. Reungwetwattana, T.; Nakagawa, K.; Cho, B.C.; Cobo, M.; Cho, E.K.; Bertolini, A.; Bohnet, S.; Zhou, C.; Lee, K.H.; Nogami, N.; et al. CNS Response to Osimertinib Versus Standard Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients with Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 3290–3297. [Google Scholar] [CrossRef]
  49. Jänne, P.A.; Planchard, D.; Kobayashi, K.; Cheng, Y.; Lee, C.K.; Valdiviezo, N.; Laktionov, K.; Yang, T.Y.; Yu, Y.; Kato, T.; et al. CNS Efficacy of Osimertinib with or Without Chemotherapy in Epidermal Growth Factor Receptor–Mutated Advanced Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2024, 42, 808–820. [Google Scholar] [CrossRef]
  50. Solomon, B.J.; Liu, G.; Felip, E.; Mok, T.S.K.; Soo, R.A.; Mazieres, J.; Shaw, A.T.; de Marinis, F.; Goto, Y.; Wu, Y.-L.; et al. Lorlatinib Versus Crizotinib in Patients with Advanced ALK-Positive Non–Small Cell Lung Cancer: 5-Year Outcomes From the Phase III CROWN Study. J. Clin. Oncol. 2024, 42, 3400–3409. [Google Scholar] [CrossRef]
  51. Powell, S.F.; Rodríguez-Abreu, D.; Langer, C.J.; Tafreshi, A.; Paz-Ares, L.; Kopp, H.-G.; Rodríguez-Cid, J.; Kowalski, D.M.; Cheng, Y.; Kurata, T.; et al. Outcomes with Pembrolizumab Plus Platinum-Based Chemotherapy for Patients with NSCLC and Stable Brain Metastases: Pooled Analysis of KEYNOTE-021, -189, and -407. J. Thorac. Oncol. 2021, 16, 1883–1892. [Google Scholar] [CrossRef]
  52. Nadal, E.; Rodríguez-Abreu, D.; Simó, M.; Massutí, B.; Juan, O.; Huidobro, G.; López, R.; De Castro, J.; Estival, A.; Mosquera, J.; et al. Phase II Trial of Atezolizumab Combined with Carboplatin and Pemetrexed for Patients with Advanced Nonsquamous Non-Small-Cell Lung Cancer with Untreated Brain Metastases (Atezo-Brain, GECP17/05). J. Clin. Oncol. 2023, 41, 4478–4485. [Google Scholar] [CrossRef]
  53. Hou, X.; Zhou, C.; Wu, G.; Lin, W.; Xie, Z.; Zhang, H.; Yi, J.; Peng, Z.; Yin, L.; Ma, C.; et al. Efficacy, Safety, and Health-Related Quality of Life with Camrelizumab Plus Pemetrexed and Carboplatin as First-Line Treatment for Advanced Nonsquamous NSCLC with Brain Metastases (CAP-BRAIN): A Multicenter, Open-Label, Single-Arm, Phase 2 Study. J. Thorac. Oncol. 2023, 18, 769–779. [Google Scholar] [CrossRef] [PubMed]
  54. Coleman, N.; Yap, T.A.; Heymach, J.V.; Meric-Bernstam, F.; Le, X. Antibody-Drug Conjugates in Lung Cancer: Dawn of a New Era? npj Precis. Oncol. 2023, 7, 5. [Google Scholar] [CrossRef] [PubMed]
  55. Ha, S.Y.; Choi, S.-J.; Cho, J.H.; Choi, H.J.; Lee, J.; Jung, K.; Irwin, D.; Liu, X.; Lira, M.E.; Mao, M.; et al. Lung Cancer in Never-Smoker Asian Females Is Driven by Oncogenic Mutations, Most Often Involving EGFR. Oncotarget 2015, 6, 5465–5474. [Google Scholar] [CrossRef] [PubMed]
  56. Li, B.T.; Planchard, D.; Goto, K.; Smit, E.F.; Langen, J.D.; Goto, Y.; Ninomiya, K.; Kubo, T.; Pérol, M.; Felip, E.; et al. 1321MO Trastuzumab Deruxtecan (T-DXd) in Patients (Pts) with HER2 (ERBB2)-Mutant (HER2m) Metastatic Non–Small Cell Lung Cancer (NSCLC) with and without Brain Metastases (BMs): Pooled Analyses from DESTINY-Lung01 and DESTINY-Lung02. Ann. Oncol. 2023, 34, S762–S763. [Google Scholar] [CrossRef]
  57. Lenárt, S.; Lenárt, P.; Šmarda, J.; Remšík, J.; Souček, K.; Beneš, P. Trop2: Jack of All Trades, Master of None. Cancers 2020, 12, 3328. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7696911/ (accessed on 23 June 2024). [CrossRef]
  58. Inamura, K.; Yokouchi, Y.; Kobayashi, M.; Ninomiya, H.; Sakakibara, R.; Subat, S.; Nagano, H.; Nomura, K.; Okumura, S.; Shibutani, T.; et al. Association of Tumor TROP2 Expression with Prognosis Varies among Lung Cancer Subtypes. Oncotarget 2017, 8, 28725. [Google Scholar] [CrossRef]
  59. Bessede, A.; Peyraud, F.; Besse, B.; Cousin, S.; Cabart, M.; Chomy, F.; Rey, C.; Lara, O.; Odin, O.; Nafia, I.; et al. TROP2 Is Associated with Primary Resistance to Immune Checkpoint Inhibition in Patients with Advanced Non-Small Cell Lung Cancer. Clin. Cancer Res. 2024, 30, 779–785. [Google Scholar] [CrossRef]
  60. Chen, Q.; Jia, G.; Zhang, X.; Ma, W. Targeting HER3 to Overcome EGFR TKI Resistance in NSCLC. Front. Immunol. 2024, 14, 1332057. [Google Scholar] [CrossRef]
  61. Gandullo-Sánchez, L.; Ocaña, A.; Pandiella, A. HER3 in Cancer: From the Bench to the Bedside. J. Exp. Clin. Cancer Res. 2022, 41, 310. [Google Scholar] [CrossRef]
  62. Gherardi, E.; Birchmeier, W.; Birchmeier, C.; Vande Woude, G. Targeting MET in Cancer: Rationale and Progress. Nat. Rev. Cancer 2012, 12, 89–103. [Google Scholar] [CrossRef]
  63. Camidge, D.R.; Bar, J.; Horinouchi, H.; Goldman, J.W.; Moiseenko, F.V.; Filippova, E.; Cicin, I.; Ciuleanu, T.E.; Daaboul, N.; Liu, C.; et al. Telisotuzumab Vedotin Monotherapy in Patients with Previously Treated C-Met–Overexpressing Non-Squamous EGFR Wildtype Advanced NSCLC: Primary Analysis of the LUMINOSITY Trial. J. Clin. Oncol. 2024, 42 (Suppl. S16), 103. [Google Scholar] [CrossRef]
  64. Barnholtz-Sloan, J.S.; Sloan, A.E.; Davis, F.G.; Vigneau, F.D.; Lai, P.; Sawaya, R.E. Incidence Proportions of Brain Metastases in Patients Diagnosed (1973 to 2001) in the Metropolitan Detroit Cancer Surveillance System. J. Clin. Oncol. 2004, 22, 2865–2872. [Google Scholar] [CrossRef] [PubMed]
  65. Kuksis, M.; Gao, Y.; Tran, W.; Hoey, C.; Kiss, A.; Komorowski, A.S.; Dhaliwal, A.J.; Sahgal, A.; Das, S.; Chan, K.K.; et al. The Incidence of Brain Metastases among Patients with Metastatic Breast Cancer: A Systematic Review and Meta-Analysis. Neuro-Oncol. 2021, 23, 894–904. [Google Scholar] [CrossRef] [PubMed]
  66. Lin, N.U.; Murthy, R.K.; Abramson, V.; Anders, C.; Bachelot, T.; Bedard, P.L.; Borges, V.; Cameron, D.; Carey, L.A.; Chien, A.J.; et al. Tucatinib vs. Placebo, Both in Combination with Trastuzumab and Capecitabine, for Previously Treated ERBB2 (HER2)-Positive Metastatic Breast Cancer in Patients with Brain Metastases: Updated Exploratory Analysis of the HER2CLIMB Randomized Clinical Trial. JAMA Oncol. 2023, 9, 197–205. [Google Scholar] [CrossRef] [PubMed]
  67. Chehade, R.; Nofech-Mozes, S.; Plotkin, A.; Fan, K.Y.; Das, S.; Sahgal, A.; Moravan, V.; Jerzak, K.J. Human Epidermal Growth Factor Receptor 2-Low Breast Cancer Brain Metastases: An Opportunity for Targeted Systemic Therapies in a High-Need Patient Population. JCO Precis. Oncol. 2024, 8, e2300487. [Google Scholar] [CrossRef]
  68. Balinda, H.U.; Kelly, W.J.; Kaklamani, V.G.; Lathrop, K.I.; Canola, M.M.; Ghamasaee, P.; Sareddy, G.R.; Michalek, J.; Gilbert, A.R.; Surapaneni, P.; et al. Sacituzumab Govitecan in Patients with Breast Cancer Brain Metastases and Recurrent Glioblastoma: A Phase 0 Window-of-Opportunity Trial. Nat. Commun. 2024, 15, 6707. [Google Scholar] [CrossRef]
  69. Pestalozzi, B.C.; Holmes, E.; de Azambuja, E.; Metzger-Filho, O.; Hogge, L.; Scullion, M.; Láng, I.; Wardley, A.; Lichinitser, M.; Sanchez, R.I.L.; et al. CNS Relapses in Patients with HER2-Positive Early Breast Cancer Who Have and Have Not Received Adjuvant Trastuzumab: A Retrospective Substudy of the HERA Trial (BIG 1-01). Lancet Oncol. 2013, 14, 244–248. [Google Scholar] [CrossRef]
  70. von Minckwitz, G.; Huang, C.-S.; Mano, M.S.; Loibl, S.; Mamounas, E.P.; Untch, M.; Wolmark, N.; Rastogi, P.; Schneeweiss, A.; Redondo, A.; et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N. Engl. J. Med. 2019, 380, 617–628. [Google Scholar] [CrossRef]
  71. Diéras, V.; Miles, D.; Verma, S.; Pegram, M.; Welslau, M.; Baselga, J.; Krop, I.E.; Blackwell, K.; Hoersch, S.; Xu, J.; 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. [Google Scholar] [CrossRef]
  72. Krop, I.E.; Lin, N.U.; Blackwell, K.; Guardino, E.; Huober, J.; Lu, M.; Miles, D.; Samant, M.; Welslau, M.; Diéras, V. Trastuzumab Emtansine (T-DM1) versus Lapatinib plus Capecitabine in Patients with HER2-Positive Metastatic Breast Cancer and Central Nervous System Metastases: A Retrospective, Exploratory Analysis in EMILIA. Ann. Oncol. 2015, 26, 113–119. [Google Scholar] [CrossRef]
  73. Krop, I.E.; Kim, S.-B.; González-Martín, A.; LoRusso, P.M.; Ferrero, J.-M.; Smitt, M.; Yu, R.; Leung, A.C.F.; Wildiers, H. TH3RESA study collaborators 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. [Google Scholar] [CrossRef]
  74. Montemurro, F.; Delaloge, S.; Barrios, C.H.; Wuerstlein, R.; Anton, A.; Brain, E.; Hatschek, T.; Kelly, C.M.; Peña-Murillo, C.; Yilmaz, M.; et al. Trastuzumab Emtansine (T-DM1) in Patients with HER2-Positive Metastatic Breast Cancer and Brain Metastases: Exploratory Final Analysis of Cohort 1 from KAMILLA, a Single-Arm Phase IIIb Clinical Trial. Ann. Oncol. 2020, 31, 1350–1358. [Google Scholar] [CrossRef]
  75. Modi, S.; Park, H.; Murthy, R.K.; Iwata, H.; Tamura, K.; Tsurutani, J.; Moreno-Aspitia, A.; Doi, T.; Sagara, Y.; Redfern, C.; et al. Antitumor Activity and Safety of Trastuzumab Deruxtecan in Patients with HER2-Low-Expressing Advanced Breast Cancer: Results from a Phase Ib Study. J. Clin. Oncol. 2020, 38, 1887–1896. [Google Scholar] [CrossRef] [PubMed]
  76. Jerusalem, G.; Park, Y.H.; Yamashita, T.; Hurvitz, S.A.; Modi, S.; Andre, F.; Krop, I.E.; Gonzàlez Farré, X.; You, B.; Saura, C.; et al. Trastuzumab Deruxtecan in HER2-Positive Metastatic Breast Cancer Patients with Brain Metastases: A DESTINY-Breast01 Subgroup Analysis. Cancer Discov. 2022, 12, 2754–2762. [Google Scholar] [CrossRef] [PubMed]
  77. Bartsch, R.; Berghoff, A.S.; Furtner, J.; Marhold, M.; Bergen, E.S.; Roider-Schur, S.; Starzer, A.M.; Forstner, H.; Rottenmanner, B.; Dieckmann, K.; et al. Trastuzumab Deruxtecan in HER2-Positive Breast Cancer with Brain Metastases: A Single-Arm, Phase 2 Trial. Nat. Med. 2022, 28, 1840–1847. [Google Scholar] [CrossRef] [PubMed]
  78. Harbeck, N.; Ciruelos, E.; Jerusalem, G.; Müller, V.; Niikura, N.; Viale, G.; Bartsch, R.; Kurzeder, C.; Higgins, M.J.; Connolly, R.M.; et al. Trastuzumab Deruxtecan in HER2-Positive Advanced Breast Cancer with or without Brain Metastases: A Phase 3b/4 Trial. Nat. Med. 2024, 30, 3717–3727. [Google Scholar] [CrossRef] [PubMed]
  79. Pérez-García, J.M.; Vaz Batista, M.; Cortez, P.; Ruiz-Borrego, M.; Cejalvo, J.M.; de la Haba-Rodriguez, J.; Garrigós, L.; Racca, F.; Servitja, S.; Blanch, S.; et al. Trastuzumab Deruxtecan in Patients with Central Nervous System Involvement from HER2-Positive Breast Cancer: The DEBBRAH Trial. Neuro-Oncol. 2023, 25, 157–166. [Google Scholar] [CrossRef]
  80. Venetis, K.; Crimini, E.; Sajjadi, E.; Corti, C.; Guerini-Rocco, E.; Viale, G.; Curigliano, G.; Criscitiello, C.; Fusco, N. HER2 Low, Ultra-Low, and Novel Complementary Biomarkers: Expanding the Spectrum of HER2 Positivity in Breast Cancer. Front. Mol. Biosci. 2022, 9, 834651. [Google Scholar] [CrossRef]
  81. Mosele, F.; Deluche, E.; Lusque, A.; Le Bescond, L.; Filleron, T.; Pradat, Y.; Ducoulombier, A.; Pistilli, B.; Bachelot, T.; Viret, F.; et al. Trastuzumab Deruxtecan in Metastatic Breast Cancer with Variable HER2 Expression: The Phase 2 DAISY Trial. Nat. Med. 2023, 29, 2110–2120. Available online: https://www.nature.com/articles/s41591-023-02478-2 (accessed on 1 September 2024). [CrossRef]
  82. Epaillard, N.; Lusque, A.; Pistilli, B.; André, F.; Bachelot, T.; Pierga, J.-Y.; Ducoulombier, A.; Jouannaud, C.; Viret, F.; Salabert, L.; et al. 260P Antitumor Activity of Trastuzumab Deruxtecan (T-DXd) in Patients with Metastatic Breast Cancer (mBC) and Brain Metastases (BMs) from DAISY Trial. Ann. Oncol. 2022, 33, S656. [Google Scholar] [CrossRef]
  83. Tsurutani, J.; Jacot, W.; Yamashita, T.; Riaz, F.; Yerushalmi, R.; Im, S.-A.; Niikura, N.; Halser-Strub, U.; Cortés, J.; Wennstig, A.-K.; et al. 388P Subgroup Analysis of Patients (Pts) with HER2-Low Metastatic Breast Cancer (mBC) with Brain Metastases (BMs) at Baseline from DESTINY-Breast04, a Randomized Phase III Study of Trastuzumab Deruxtecan (T-DXd) vs. Treatment of Physician’s Choice (TPC). Ann. Oncol. 2023, 34, S342–S343. [Google Scholar] [CrossRef]
  84. Batista, M.V.; Pérez-García, J.M.; Cortez, P.; Garrigós, L.; Fernández-Abad, M.; Gion, M.; Martínez-Bueno, A.; Saavedra, C.; Teruel, I.; Fernandez-Ortega, A.; et al. Trastuzumab Deruxtecan in Patients with Previously Treated HER2-Low Advanced Breast Cancer and Active Brain Metastases: The DEBBRAH Trial. ESMO Open 2024, 9, 103699. [Google Scholar] [CrossRef] [PubMed]
  85. Bardia, A.; Hurvitz, S.A.; Tolaney, S.M.; Loirat, D.; Punie, K.; Oliveira, M.; Brufsky, A.; Sardesai, S.D.; Kalinsky, K.; Zelnak, A.B.; et al. Sacituzumab Govitecan in Metastatic Triple-Negative Breast Cancer. N. Engl. J. Med. 2021, 384, 1529–1541. [Google Scholar] [CrossRef] [PubMed]
  86. Hurvitz, S.A.; Bardia, A.; Punie, K.; Kalinsky, K.; Carey, L.A.; Rugo, H.S.; Diéras, V.; Phan, S.; Delaney, R.; Zhu, Y.; et al. Subgroup Analyses from the Phase 3 ASCENT Study of Sacituzumab Govitecan in Metastatic Triple-Negative Breast Cancer. npj Breast Cancer 2024, 10, 33. [Google Scholar] [CrossRef]
  87. Terasima, T.; Tolmach, L.J. Variations in Several Responses of HeLa Cells to X-Irradiation during the Division Cycle. Biophys. J. 1963, 3, 11–33. [Google Scholar] [CrossRef]
  88. Borges, G.S.; Rovere, R.K.; Dias, S.M.K.; Chong, F.H.; Morais, M.D.S. Safety and Efficacy of the Combination of T-DM1 with Radiotherapy of the Central Nervous System in a Patient with HER2-Positive Metastatic Breast Cancer: Case Study and Review of the Literature. Ecancermedicalscience 2015, 9, 586. [Google Scholar] [CrossRef]
  89. Ricciardi, G.R.R.; Russo, A.; Franchina, T.; Schifano, S.; Mastroeni, G.; Santacaterina, A.; Adamo, V. Efficacy of T-DM1 for Leptomeningeal and Brain Metastases in a HER2 Positive Metastatic Breast Cancer Patient: New Directions for Systemic Therapy—A Case Report and Literature Review. BMC Cancer 2018, 18, 97. [Google Scholar] [CrossRef]
  90. Orth, M.; Unger, K.; Schoetz, U.; Belka, C.; Lauber, K. Taxane-Mediated Radiosensitization Derives from Chromosomal Missegregation on Tripolar Mitotic Spindles Orchestrated by AURKA and TPX2. Oncogene 2018, 37, 52–62. [Google Scholar] [CrossRef]
  91. Mignot, F.; Kirova, Y.; Verrelle, P.; Teulade-Fichou, M.-P.; Megnin-Chanet, F. In Vitro Effects of Trastuzumab Emtansine (T-DM1) and Concurrent Irradiation on HER2-Positive Breast Cancer Cells. Cancer Radiother. 2021, 25, 126–134. [Google Scholar] [CrossRef]
  92. Mitsuya, K.; Watanabe, J.; Nakasu, Y.; Hayashi, N.; Harada, H.; Ito, I. Expansive Hematoma in Delayed Cerebral Radiation Necrosis in Patients Treated with T-DM1: A Report of Two Cases. BMC Cancer 2016, 16, 391. [Google Scholar] [CrossRef]
  93. Adams, S.R.; Yang, H.C.; Savariar, E.N.; Aguilera, J.; Crisp, J.L.; Jones, K.A.; Whitney, M.A.; Lippman, S.M.; Cohen, E.E.W.; Tsien, R.Y.; et al. Anti-Tubulin Drugs Conjugated to Anti-ErbB Antibodies Selectively Radiosensitize. Nat. Commun. 2016, 7, 13019. [Google Scholar] [CrossRef] [PubMed]
  94. Vilela, M.D.; Longstreth, W.T.; Pedrosa, H.A.S.; Gil, G.O.B.; Duarte, J.M.; Filho, M.A.D. Progressively Enlarging Cerebellar Hematoma Concurrent with T-DM1 Treatment. World Neurosurg. 2018, 111, 109–114. [Google Scholar] [CrossRef] [PubMed]
  95. Carlson, J.A.; Nooruddin, Z.; Rusthoven, C.; Elias, A.; Borges, V.F.; Diamond, J.R.; Kavanagh, B.; Kabos, P. Trastuzumab Emtansine and Stereotactic Radiosurgery: An Unexpected Increase in Clinically Significant Brain Edema. Neuro-Oncol. 2014, 16, 1006–1009. [Google Scholar] [CrossRef]
  96. Crone, S.A.; Zhao, Y.-Y.; Fan, L.; Gu, Y.; Minamisawa, S.; Liu, Y.; Peterson, K.L.; Chen, J.; Kahn, R.; Condorelli, G.; et al. ErbB2 Is Essential in the Prevention of Dilated Cardiomyopathy. Nat. Med. 2002, 8, 459–465. [Google Scholar] [CrossRef]
  97. Vartanian, T.; Goodearl, A.; Viehöver, A.; Fischbach, G. Axonal Neuregulin Signals Cells of the Oligodendrocyte Lineage through Activation of HER4 and Schwann Cells through HER2 and HER3. J. Cell Biol. 1997, 137, 211–220. [Google Scholar] [CrossRef]
  98. Lebow, E.S.; Pike, L.R.G.; Seidman, A.D.; Moss, N.; Beal, K.; Yu, Y. Symptomatic Necrosis with Antibody-Drug Conjugates and Concurrent Stereotactic Radiotherapy for Brain Metastases. JAMA Oncol. 2023, 9, 1729–1733. [Google Scholar] [CrossRef]
  99. Lee, E.Q.; Camidge, D.R.; Mehta, G. Extending Our Reach: Expanding Enrollment in Brain Metastases and Primary Brain Tumor Clinical Trials. Am. Soc. Clin. Oncol. Educ. Book. 2022, 42, 1–9. [Google Scholar] [CrossRef] [PubMed]
  100. Rahman, R.; Polley, M.-Y.C.; Alder, L.; Brastianos, P.K.; Anders, C.K.; Tawbi, H.A.; Mehta, M.; Wen, P.Y.; Geyer, S.; de Groot, J.; et al. Current Drug Development and Trial Designs in Neuro-Oncology: Report from the First American Society of Clinical Oncology and Society for Neuro-Oncology Clinical Trials Conference. Lancet Oncol. 2023, 24, e161–e171. [Google Scholar] [CrossRef]
  101. Sivakumar, S.; Jin, D.X.; Tukachinsky, H.; Murugesan, K.; McGregor, K.; Danziger, N.; Pavlick, D.; Gjoerup, O.; Ross, J.S.; Harmon, R.; et al. Tissue and Liquid Biopsy Profiling Reveal Convergent Tumor Evolution and Therapy Evasion in Breast Cancer. Nat. Commun. 2022, 13, 7495. [Google Scholar] [CrossRef]
  102. Ogitani, Y.; Aida, T.; Hagihara, K.; Yamaguchi, J.; Ishii, C.; Harada, N.; Soma, M.; Okamoto, H.; Oitate, M.; Arakawa, S.; et al. DS-8201a, A Novel HER2-Targeting ADC with a Novel DNA Topoisomerase I Inhibitor, Demonstrates a Promising Antitumor Efficacy with Differentiation from T-DM1. Clin. Cancer Res. 2016, 22, 5097–5108. [Google Scholar] [CrossRef]
  103. Ji, C.; Li, F.; Yuan, Y.; Zhang, H.; Bian, L.; Zhang, S.; Wang, T.; Li, J.; Jiang, Z. Novel Anti-HER2 Antibody-Drug Conjugates Versus T-DM1 for HER2-Positive Metastatic Breast Cancer After Tyrosine Kinase Inhibitors Treatment. Oncologist 2023, 28, e859–e866. [Google Scholar] [CrossRef] [PubMed]
  104. Jain, S.; Griffith, J.I.; Porath, K.A.; Rathi, S.; Le, J.; Pasa, T.I.; Decker, P.A.; Gupta, S.K.; Hu, Z.; Carlson, B.L.; et al. Bystander Effects, Pharmacokinetics, and Linker-Payload Stability of EGFR-Targeting Antibody-Drug Conjugates Losatuxizumab Vedotin and Depatux-M in Glioblastoma Models. Clin. Cancer Res. 2024, 30, 3287–3297. [Google Scholar] [CrossRef] [PubMed]
  105. Whalen, K.A.; White, B.H.; Quinn, J.M.; Kriksciukaite, K.; Alargova, R.; Au Yeung, T.P.; Bazinet, P.; Brockman, A.; DuPont, M.M.; Oller, H.; et al. Targeting the Somatostatin Receptor 2 with the Miniaturized Drug Conjugate, PEN-221: A Potent and Novel Therapeutic for the Treatment of Small Cell Lung Cancer. Mol. Cancer Ther. 2019, 18, 1926–1936. [Google Scholar] [CrossRef]
  106. Anami, Y.; Otani, Y.; Xiong, W.; Ha, S.Y.Y.; Yamaguchi, A.; Rivera-Caraballo, K.A.; Zhang, N.; An, Z.; Kaur, B.; Tsuchikama, K. Homogeneity of Antibody-Drug Conjugates Critically Impacts the Therapeutic Efficacy in Brain Tumors. Cell Rep. 2022, 39, 110839. [Google Scholar] [CrossRef]
  107. Nguyen, T.D.; Bordeau, B.M.; Balthasar, J.P. Mechanisms of ADC Toxicity and Strategies to Increase ADC Tolerability. Cancers 2023, 15, 713. [Google Scholar] [CrossRef]
  108. Kaempffe, A.; Dickgiesser, S.; Rasche, N.; Paoletti, A.; Bertotti, E.; De Salve, I.; Sirtori, F.R.; Kellner, R.; Könning, D.; Hecht, S.; et al. Effect of Conjugation Site and Technique on the Stability and Pharmacokinetics of Antibody-Drug Conjugates. J. Pharm. Sci. 2021, 110, 3776–3785. [Google Scholar] [CrossRef] [PubMed]
  109. El Alaoui, M.; Sivado, E.; Jallas, A.-C.; Mebarki, L.; Dyson, M.R.; Perrez, F.; Valsesia-Wittmann, S.; El Alaoui, S. Antibody and Antibody Fragments Site-Specific Conjugation Using New Q-Tag Substrate of Bacterial Transglutaminase. Cell Death Discov. 2024, 10, 79. [Google Scholar] [CrossRef]
  110. Matsuda, Y.; Shikida, N.; Hatada, N.; Yamada, K.; Seki, T.; Nakahara, Y.; Endo, Y.; Shimbo, K.; Takahashi, K.; Nakayama, A.; et al. AJICAP-M: Traceless Affinity Peptide Mediated Conjugation Technology for Site-Selective Antibody-Drug Conjugate Synthesis. Org. Lett. 2024, 26, 5597–5601. [Google Scholar] [CrossRef]
  111. Mair, M.J.; Bartsch, R.; Le Rhun, E.; Berghoff, A.S.; Brastianos, P.K.; Cortes, J.; Gan, H.K.; Lin, N.U.; Lassman, A.B.; Wen, P.Y.; et al. Understanding the Activity of Antibody-Drug Conjugates in Primary and Secondary Brain Tumours. Nat. Rev. Clin. Oncol. 2023, 20, 372–389. [Google Scholar] [CrossRef]
  112. Ballestín, P.; López de Sá, A.; Díaz-Tejeiro, C.; Paniagua-Herranz, L.; Sanvicente, A.; López-Cade, I.; Pérez-Segura, P.; Alonso-Moreno, C.; Nieto-Jiménez, C.; Ocaña, A. Understanding the Toxicity Profile of Approved ADCs. Pharmaceutics 2025, 17, 258. [Google Scholar] [CrossRef]
  113. Watanabe, T.; Arashida, N.; Fujii, T.; Shikida, N.; Ito, K.; Shimbo, K.; Seki, T.; Iwai, Y.; Hirama, R.; Hatada, N.; et al. Exo-Cleavable Linkers: Enhanced Stability and Therapeutic Efficacy in Antibody–Drug Conjugates. J. Med. Chem. 2024, 67, 18124–18138. [Google Scholar] [CrossRef] [PubMed]
  114. Chuprakov, S.; Ogunkoya, A.O.; Barfield, R.M.; Bauzon, M.; Hickle, C.; Kim, Y.C.; Yeo, D.; Zhang, F.; Rabuka, D.; Drake, P.M. Tandem-Cleavage Linkers Improve the In Vivo Stability and Tolerability of Antibody–Drug Conjugates. Bioconjugate Chem. 2021, 32, 746–754. [Google Scholar] [CrossRef] [PubMed]
  115. Vaz Batista, M.; Pérez-García, J.M.; Garrigós, L.; García-Sáenz, J.Á.; Cortez, P.; Racca, F.; Blanch, S.; Ruiz-Borrego, M.; Fernández-Ortega, A.; Fernández-Abad, M.; et al. The DEBBRAH Trial: Trastuzumab Deruxtecan in HER2-Positive and HER2-Low Breast Cancer Patients with Leptomeningeal Carcinomatosis. Med 2025, 6, 100502. [Google Scholar] [CrossRef] [PubMed]
  116. Murthy, R.K.; Loi, S.; Okines, A.; Paplomata, E.; Hamilton, E.; Hurvitz, S.A.; Lin, N.U.; Borges, V.; Abramson, V.; Anders, C.; et al. Tucatinib, Trastuzumab, and Capecitabine for HER2-Positive Metastatic Breast Cancer. N. Engl. J. Med. 2020, 382, 597–609. [Google Scholar] [CrossRef]
Figure 1. Mechanism of action of ADCs. Abbreviations: ADC, antibody–drug conjugate.
Figure 1. Mechanism of action of ADCs. Abbreviations: ADC, antibody–drug conjugate.
Cancers 17 01591 g001
Table 1. FDA-approved ADCs for solid tumors.
Table 1. FDA-approved ADCs for solid tumors.
ADC Target Linker Type Payload Indications Registration Trial
Mirvetuximab
soravtansine
Folate receptor alpha Cleavable Maytansinoid DM4 Folate receptor-alpha (FRα)-positive, platinum-resistant high-grade serous epithelial ovarian, fallopian tube, or primary peritoneal cancer patients who have received 1 to 3 prior systemic treatment regimens SORAYA (NCT04296890)
Tisotumab
vedotin
Tissue Factor Cleavable Monomethyl
Auristatin
E (MMAE)
Recurrent or metastatic cervical cancer with disease progression to chemotherapy InnovaTV 301 (NCT04697628)
Ado-trastuzumab emtansine HER2 Non
cleavable
Maytansine DM1 EBC:
Adjuvant treatment of patients with HER2-positive EBC who have residual invasive disease after neoadjuvant taxane- and trastuzumab-based treatment.
MBC:
Patients with HER2-positive MBC who previously received trastuzumab and a taxane, separately or in combination
KATHERINE (NCT01772472) EMILIA (NCT00829166)
Enfortumab
vedotin
Nectin-4 Clevable Monomethyl
Auristatin
E (MMAE)
Adult patients with locally advanced or metastatic urothelial cancer who have received an anti PD-(L)1 agent, and platinum-based chemotherapy EV-301 (NCT03474107)
Trastuzumab
deruxtecan
HER2 Cleavable Deruxtecan Breast cancer:
Unresectable or metastatic HER2-positive
(IHC 3+ or ISH positive) BC patients who have received a prior anti-HER2-based regimen
Patients with unresectable or metastatic HER2-low
(IHC 1+ or IHC 2+/ISH−) BC, as determined by an FDA-approved test, who have received a prior chemotherapy in the metastatic setting
Non-small cell lung cancer:
Unresectable or metastatic NSCLC patients whose tumors have activating HER2 mutations, as detected by an FDA-approved test, and who have received a prior systemic therapy (accelerated approval)
Gastric cancer:
Locally advanced or metastatic HER2-positive (IHC 3+ or IHC 2+/ISH positive) gastric or gastroesophageal junction (GEJ) adenocarcinoma patients who have received a prior trastuzumab-based
regimen
Solid tumors: Patients with unresectable or metastatic HER2-positive (IHC3+) solid tumors who have received prior systemic treatment (accelerated approval)
DESTINY-Breast03 (NCT03529110) DESTINY-Breast04 (NCT03734029) DESTINY-Lung02 (NCT04644237) DESTINY-Gastric01 (NCT03329690) DESTINY-PanTumor02 (NCT04482309)
Sacituzumab
Govitecan
TROP2 Cleavable Camptothecin analog (SN38) BC:
Unresectable locally advanced or metastatic triple-negative BC patients who have received 2 or more prior systemic therapies
Urothelial cancer:
Locally advanced or metastatic urothelial cancer (mUC) patients who have previously received platinum-based chemotherapy and an anti PD-(L)1 inhibitor. (accelerated approval)
ASCENT (NCT02574455) IMMU-132-01 (NCT01631552) TROPHY (NCT03547973)
Datopotomab DeruxtecanTROP2CleavableTopoisomerase I inhibitor
(Deruxtecan)
Patients with unresectable or metastatic hormone receptor-positive, HER2-breast cancer that has progressed after endocrine-based therapy and chemotherapy for unresectable or metastatic diseaseTROPION-Breast01 (NCT05104866)
Abbreviations: ADC, antibody–drug conjugates; BC, breast cancer; DM1, Maytansinoid Derivative 1; EBC, Early breast cancer; DM4, Maytansinoid Derivative 4; EBC, early breast cancer; FRα, folate receptor-alpha; HER2, human epidermal growth factor receptor 2; IHC, immunohistochemistry; ISH, in situ hybridization; MBC, metastatic breast cancer; MMAE, monomethyl auristatin E; NCT, National Clinical Trial; NSCLC, non-small cell lung cancer; PD-1, Programmed Death-1; PD-L1, Programmed Death-Ligand 1; SN38, Camptothecin analog, the active metabolite of irinotecan.
Table 3. Summary of ongoing clinical trial evaluating ADC in primary brain tumors patients.
Table 3. Summary of ongoing clinical trial evaluating ADC in primary brain tumors patients.
Clinical Trial Response Endpoint Treatment Target Linker Type Payload
Action
Population Trial Type Status
AMG 595 First-in-Human in Recurrent Gliomas
(NCT01475006)
ORR by Macdonald criteria AMG 595 EGFRvIII Non-Cleavable Microtubule
inhibitor (Maytansinoid DM1)
Patients with rGBM (Part 1 and 2) or WHO Grade III anaplastic astrocytoma (Part 1 only) expressing EGFRvIII Phase I Completed. No results published (last update 2016)
Safety and Efficacy of
L19TNF in Patients with Isocitrate Dehydrogenase (IDH) Wildtype WHO Grade III/IV Glioma at First Relapse (GLIOMOON)
(NCT03779230)
PFS, OS, ORR by iRANO criteria Onfekafusp alfa (L19TNF) Fibronectin NA TNFalpha Patients with WHO Grade III/IV Glioma at First Relapse Phase I/II Completed
Safety and Efficacy of L19TNF Plus Temozolomide Chemoradiotherapy in Patients with Newly Diagnosed Glioblastoma (GLIOSUN)
(NCT04443010)
OS, PFS, ORR, DCR, BORR by iRANO Onfekafusp alfa (L19TNF) + TMZ Fibronectin NA TNFalpha Patients with newly diagnosed GBM Phase II Recruiting
A Study to Evaluate Safety and Efficacy of L19TNF Plus Lomustine in Patients with Glioblastoma at First Progression (GLIOSTAR)
(NCT04573192)
OS, PFS by iRANO Onfekafusp alfa (L19TNF) + lomustine Fibronectin NA TNFalpha Patients with GBM at first progression Phase I/II Recruiting
Neuro/Sacituzumab Govitecan/Breast Brain Metastasis/Glioblastoma/Ph 0
(NCT03995706)
Ratio of SN-38 and its metabolites relative to serum concentration Sacituzumab GovitecanTROP2 Cleavable Topoisomerase I
Inhibitor (govitecan)
Patients with rGBM/BM from breast Phase 0 Completed
Abbreviations: ADC, antibody–drug conjugate; BM, brain metastasis; BORR, best overall response rate; DCR, disease control rate; EGFRvIII, epidermal growth factor receptor variant III; iRANO, Immunotherapy Response Assessment in Neuro-Oncology; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; TNFalpha, tumor necrosis factor alfa; TROP2, trophoblast cell surface antigen 2.
Table 4. Summary of available clinical trials of ADCs in NSCLC with data on intracranial activity.
Table 4. Summary of available clinical trials of ADCs in NSCLC with data on intracranial activity.
Clinical Trial Type of Trial ADC Target Linker Type Payload Action Population CNS Metastasis
Eligibility
Intracranial Activity Intracranic Response Criteria
TROPION-Lung05 (NCT04484142) Phase IIDatopotamab Deruxtecan (Dato-DXd) TROP-2 Cleavable Topoisomerase I inhibitor
(Deruxtecan)
a/m NSCLC with AGAs who progressed following targeted therapy and platinum-based CT Clinically inactive BM (not requiring medical therapy) eligible. Leptomeningeal disease excluded iORR 22% iDCR 72% RECIST v1.1
DESTINY-Lung01 (NCT03505710)
DESTINY-Lung02 (NCT04644237)
Phase IITrastuzumab-Deruxtecan (T-DXd) HER2 Cleavable Topoisomerase I inhibitor
(Deruxtecan)
HER2-overexpressing or HER2-mutant unresectable or metastatic NSCLC No exclusion criteria for patients with brain metastases, including leptomeningeal carcinomatosis 5.4 mg/kg Cohort (DL-02): iORR 25%
iDCR 81.3% 6.4 mg/kg Cohort (DL-01/02): iORR 18.5%
iDCR 76%
RECIST v1.1
HERTHENA-Lung01 (NCT04619004) Phase IIPatritumab Deruxtecan (HER3-DXd) HER3 Cleavable Topoisomerase I inhibitor
(Deruxtecan)
a/m NSCLC with canonical EGFR mutation who progressed on at least one EGFR TKI and one platinum-based CT Clinically inactive BM (not requiring medical therapy) eligible. Leptomeningeal disease excluded iORR 33.3% iDCR 76.6% RECIST v1.1
Abbreviations: a/m, advanced/metastatic; ADC, antibody–drug conjugate; AGAs, actionable genomic alterations; CT, chemotherapy; Dxd, Deruxtecan; HER2, human epidermal growth factor receptor 2; HER3, human epidermal growth factor receptor 3; iDCR, intracranial disease control rate; iORR, intracranial overall response rate; NSCLC, non-small cell lung cancer; TROP-2, trophoblast cell-surface antigen 2.
Table 5. Summary of available clinical trials of ADCs in BC with data on intracranial activity.
Table 5. Summary of available clinical trials of ADCs in BC with data on intracranial activity.
Clinical Trial Type of Trial ADC Target Linker Type Payload Action Population CNS Metastasis
Eligibility
CNS Endpoints Intracranic Response Criteria
EMILIA (NCT008291) Retrospective
exploratory analysis
Trastuzumab emtansine (T-DM1) HER2 Non-cleavable Microtubule
Inhibitor (Maytansinoid DM1)
HER2-positive a/m BC, previously treated with trastuzumab and a taxane Treated, asymptomatic Patients with baseline BM mOS: 26.8 vs. 12.9 mo RECIST v1.1
TH3RESA
(NCT014191)
Phase III Trastuzumab emtansine (T-DM1) HER2 Non-cleavable Microtubule
Inhibitor (Maytansinoid DM1)
HER2-positive mBC pretreated with ≥2 HER2 directed regimens Treated, asymptomatic Patients with baseline BM mPFS: 5.8 vs. 2.9 mo RECIST v1.1
KAMILLA (NCT017025) Exploratory analysis Trastuzumab emtansine (T-DM1) HER2 Non-cleavable Microtubule
Inhibitor (Maytansinoid DM1)
HER2-positive a/m BC progressing after CT and anti-HER2 therapy or ≤6 mo after adjuvant therapy Controlled or untreated and asymptomatic iORR: 21.4%, iCR: 2.4% RECIST v1.1
DESTINY-Breast01
(NCT032484)
Phase II Trastuzumab
Deruxtecan (T-DXd)
HER2 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
HER2-positive mBC previously treated with T-DM1 Treated, asymptomatic iORR: 41.2% RECIST v1.1
DESTINY-Breast03 (NCT035291) Phase III Trastuzumab
Deruxtecan (T-DXd)
HER2 Cleavable Topoisomerase I Inhibitor (Deruxtecan) HER2-positive mBC previously treated with trastuzumab and a taxane Treated or asymptomatic iORR: 79 vs. 35% RECIST v1.1
TUXEDO-1 (NCT047520) Phase II Trastuzumab
Deruxtecan (T-DXd)
HER2 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
HER2-positive mBC and newly diagnosed untreated BM/BM progressing after previous local therapy, previous exposure to trastuzumab and pertuzumab and no indication for immediate local therapy Newly diagnosed untreated/progressing after previous local therapy iORR: 73.3% iCR: 13.3% iPR: 60% iSD: 20% RANO-BM
DESTINY-Breast12 (NCT047397) Phase IIIb/IV Trastuzumab- Deruxtecan (T-DXd) HER2 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
HER2-positive mBC treated with one or more prior anti-HER2–based regimens Stable or active (untreated/previously treated and progressing) iORR: 71.7% (79.2% with stable BM
and 62.3% with active BM)
RECIST v1.1
DEBBRAH (NCT044205) Phase II Trastuzumab- Deruxtecan (T-DXd) HER2 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
HER2-positive or HER-2 low a/m BC pretreated Cohort 2: asymptomatic, untreated
Cohort 3: progressing after local therapy
CNS ORR: 50% (cohort 2) 44.4% (cohort 3) RANO-BM
ASCENT (NCT025744) Subgroup analysis Sacituzumab Govitecan (SG) TROP2 Cleavable Topoisomerase I Inhibitor (govitecan) relapsed or refractory mTNBC Stable mOS in BM population:
6.8 vs. 7.5 mo
RECIST v1.1
Abbreviations: a/m, advanced/metastatic; ADC, antibody–drug conjugate; BC, breast cancer; BM, brain metastases; CT, chemotherapy; Dxd, Deruxtecan; HER2, human epidermal growth factor receptor 2; iORR, intracranial overall response rate; iCR, intracranial complete response; iPR, intracranial partial response; iSD, intracranial stable disease; mOS, median overall survival; mPFS, median progression-free survival; mTNBC, metastatic triple-negative breast cancer; mo, months; TROP2, trophoblast cell-surface antigen 2.
Table 6. Summary of ongoing clinical trials evaluating ADC in mBC patients with BM.
Table 6. Summary of ongoing clinical trials evaluating ADC in mBC patients with BM.
Clinical Trial Response Endpoint ADC Target Linker Type Payload Action Population CNS Metastasis
Eligibility
Trial Type Status
DATO-BASE: DATOpotamab-deruxtecan for Breast Cancer Brain MetAstaSEs (NCT06176261) iORR as defined by RANO-BM criteria Datopotamab Deruxtecan (Dato-DXd) TROP2 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
HER2-negative mBC Cohorts A and B: newly diagnosed or progressing after local and/or systemic therapy Cohort C: leptomeningeal disease Phase II Recruiting
T-DXd Therapy for HER2-low Breast Cancer Patients with Brain Metastases (TUXEDO-4) (NCT06048718) iORR as defined by RANO-BM criteria Trastuzumab Deruxtecan (T-DXd) HER2 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
HER2-low mBC with newly diagnosed or progressing Bm with or without untreated type II LMD, who received ≥1 line of systemic treatment in the advanced setting Newly diagnosed or progressive, without indication for immediate local therapyPhase II Recruiting
Testing Sacituzumab Govitecan Therapy in Patients With HER2-Negative Breast Cancer and Brain Metastases (NCT04647916) iORR as defined by RANO-BM criteria Sacituzumab
govitecan (SG)
TROP2Cleavable Topoisomerase I Inhibitor (govitecan) HER2-negative mBC with BM ≥1 BM not previously treated or progressed to RT Phase II Recruiting
HER3-DXd in Breast Cancer and NSCLC Brain Metastases and Solid Tumor Leptomeningeal Disease (TUXEDO-3) (NCT05865990) iORR as defined by RANO-BM criteria Patritumab
deruxtecan (HER3-DXd)
HER3 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
mBC or aNSCLC with active BM), who received ≥1 line of systemic treatment in the advanced setting or with active LMD after RT from an advanced solid tumor who do not need immediate local treatment, and have not received prior treatment with an anti-HER3 Active BM from MBC (Cohort 1) and aNSCLC (Cohort 2), untreated LMD (Cohort 3) Phase II Active, not recruiting
Patritumab Deruxtecan (U3-1402) in Unresectable Locally Advanced or Metastatic Breast Cancer (ICARUS BREAST)
(NCT04965766)
ORRas defined by RECIST Patritumab
deruxtecan (HER3-DXd)
HER3 Clevable Topoisomerase I
Inhibitor (Deruxtecan)
HR+, ABC, resistant to ET and CDK4/6i, that must have received only one line of chemotherapy for ABC Clinically inactive, treated, asymptomatic Phase II Active, not recruiting
A Phase 1b/2 Study of T-DXd Combinations in HER2-positive Metastatic Breast Cancer (DESTINY-Breast07) (NCT04538742) ORR, PFS, PFS2, DoR as defined by RECIST, OS Trastuzumab
Deruxtecan (T-DXd)
HER2 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
HER2-positive a/mBC, ≥1 line of systemic treatment in the advanced setting (Part 1), no prior lines for a/mBC (Part 2 Modules 0–5), zero or one prior lines for a/mBC allowed (Part 2 Modules 6 and 7 Stable (Modules 0–5);
untreated, not needing local therapy or previously treated that have progressed since prior local therapy (Module 6 and 7) (<2 mg dexamethasone/day)
Phase Ib/II Active, not recruiting
PRE-I-SPY Phase I/Ib Oncology Platform Program
(PRE-I-SPY-PI) (NCT05868226)
ORR, DoR, PFS, CBR as defined by RECIST Trastuzumab
Deruxtecan (T-DXd)
HER2 Cleavable Topoisomerase I
Inhibitor (Deruxtecan)
HER2 expressing (>HER2 1+) mBCTreated and clinically stable Phase I/Ib Recruiting
Abbreviations: ABC, advanced breast cancer; ADC, antibody–drug conjugate; a/m, advanced/metastatic; aNSCLC, advanced non-small cell lung cancer; BM, brain metastasis; CBR, clinical benefit rate; CDK4/6i, cyclin-dependent kinases 4 and 6 inhibitors; DoR, duration of response; Dxd, Deruxtecan; ET, endocrine therapy; HER2, human epidermal growth factor receptor 2; HER3, human epidermal growth factor receptor 3; HR+, hormone receptor-positive; iORR, intracranial overall response rate; LMD, leptomeningeal disease; OS, overall survival; PFS, progression-free survival; RT, radiotherapy; TROP2, trophoblast cell-surface antigen 2.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bruzzone, F.; Barigazzi, C.; Di Muzio, A.; Tallarico, I.; Dipasquale, A.; Losurdo, A.; Persico, P.; Navarria, P.; Pessina, F.; Santoro, A.; et al. Exploring the Role of ADCs in Brain Metastases and Primary Brain Tumors: Insight and Future Directions. Cancers 2025, 17, 1591. https://doi.org/10.3390/cancers17091591

AMA Style

Bruzzone F, Barigazzi C, Di Muzio A, Tallarico I, Dipasquale A, Losurdo A, Persico P, Navarria P, Pessina F, Santoro A, et al. Exploring the Role of ADCs in Brain Metastases and Primary Brain Tumors: Insight and Future Directions. Cancers. 2025; 17(9):1591. https://doi.org/10.3390/cancers17091591

Chicago/Turabian Style

Bruzzone, Francesco, Chiara Barigazzi, Antonio Di Muzio, Isabel Tallarico, Angelo Dipasquale, Agnese Losurdo, Pasquale Persico, Pierina Navarria, Federico Pessina, Armando Santoro, and et al. 2025. "Exploring the Role of ADCs in Brain Metastases and Primary Brain Tumors: Insight and Future Directions" Cancers 17, no. 9: 1591. https://doi.org/10.3390/cancers17091591

APA Style

Bruzzone, F., Barigazzi, C., Di Muzio, A., Tallarico, I., Dipasquale, A., Losurdo, A., Persico, P., Navarria, P., Pessina, F., Santoro, A., & Simonelli, M. (2025). Exploring the Role of ADCs in Brain Metastases and Primary Brain Tumors: Insight and Future Directions. Cancers, 17(9), 1591. https://doi.org/10.3390/cancers17091591

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop