Next Article in Journal
The Antagonistic and Synergistic Role of Fe3+ Compounds in Chemo- and Electrochemotherapy in Human Colon Cancer In Vitro
Previous Article in Journal
New Thiazolidine-4-One Derivatives as SARS-CoV-2 Main Protease Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Progress and Innovative Combination Therapies in Trop-2-Targeted ADCs

Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(5), 652; https://doi.org/10.3390/ph17050652
Submission received: 28 April 2024 / Revised: 12 May 2024 / Accepted: 15 May 2024 / Published: 17 May 2024
(This article belongs to the Section Pharmacology)

Abstract

:
Precise targeting has become the main direction of anti-cancer drug development. Trophoblast cell surface antigen 2 (Trop-2) is highly expressed in different solid tumors but rarely in normal tissues, rendering it an attractive target. Trop-2-targeted antibody-drug conjugates (ADCs) have displayed promising efficacy in treating diverse solid tumors, especially breast cancer and urothelial carcinoma. However, their clinical application is still limited by insufficient efficacy, excessive toxicity, and the lack of biological markers related to effectiveness. This review summarizes the clinical trials and combination therapy strategies for Trop-2-targeted ADCs, discusses the current challenges, and provides new insights for future advancements.

1. Introduction

Cancer remains a crucial driver of worldwide mortality, with an estimated 20 million new diagnoses and 10 million fatalities in 2022 [1]. For decades, cytotoxic agent-based chemotherapy has been the predominant treatment modality for a diverse range of cancers [2]. However, a majority of these chemotherapy agents have a narrow therapeutic window, attributed to substantial off-target tissue exposure and consequent severe adverse effects [3]. Therefore, targeted therapy has emerged as a promising strategy for cancer treatment, attributed to its demonstrated efficacy and favorable safety profile [4]. Antibody–drug conjugates (ADCs), as a type of targeted therapy, accurately pinpoint tumor cells by connecting specific monoclonal antibodies with powerful chemotherapy drugs through stable linkers [5]. In comparison to traditional chemotherapy, ADCs offer increased tumor selectivity, enhanced efficacy, and reduced toxic side effects and have garnered significant attention in recent research [6]. Reaching a growing array of targets and indications, ADCs are leading the path towards a fresh era in precise cancer therapy [7].
Trophoblast cell surface antigen 2 (Trop-2), a type I transmembrane glycoprotein, is overexpressed in various epithelium-derived tumors (Figure 1) and is strongly associated with malignant behavior. Consequently, it has been identified as an appealing target for cancer therapy [8]. The approval of the first Trop-2-targeted ADC, IMMU-132, in April 2020 has ushered in new possibilities in cancer treatment [9]. The success of IMMU-132 has raised the hope for new treatments for patients suffering from refractory cancer. This review provides a comprehensive overview of drug development, combination therapy, and potential enhancement strategies in Trop-2-targeted ADCs.

2. Trop-2

Trophoblast cell surface antigen 2 (Trop-2) is encoded by the TACSTD2 gene found on chromosome 1p32 [10,11]. This 36-kDa type I transmembrane glycoprotein is made up of 323 amino acids and consists of various sections: a hydrophobic signal peptide (amino acids 1 to 26), an extracellular domain (amino acids 27 to 274), a transmembrane domain (amino acids 275 to 297), and a cytoplasmic tail (amino acids 298 to 323, which interact with the antibody). The extracellular domain has four N-glycosylation locations and features sequences found in the epidermal growth factor-like (EGF-like) region, with a rich cysteine area, a thyroglobulin-1 area, and a cysteine-sparse area [12]. Trop-2 is involved in regulating the cell cycle and is linked to proliferation and differentiation. Its mechanisms of action involve increasing Ki-67 expression, promoting the expression of cyclins D and E, mobilizing intracellular calcium stores to stimulate cell proliferation, and directly activating the MAPK signaling pathway and downstream AP-1 to foster cell differentiation and proliferation [13]. TROP2 overexpression not only supports tumorigenic cell proliferation, differentiation, and metastasis through pathways like JAK/STAT, Bax, Bcl-2, ERK, and Akt but also enhances PARP1 expression, leading to increased DNA replication and damage accumulation. Furthermore, the activation of Trop-2’s intracellular domain (ICD) can drive tumor cell proliferation; induce epithelial–mesenchymal transition (EMT); and boost tumor cell migration, invasion, and anti-apoptotic capabilities, ultimately hastening tumor progression [14].

3. Trop-2-Targeted ADC

3.1. Development of Trop-2-Targeted ADCs

Antibody–drug conjugates (ADCs) consist of human monoclonal antibodies (mAbs), linkers, and cytotoxic agents [7]. The mAbs specifically target antigens on the cell membrane of the desired cells and undergo internalization via endocytosis (Figure 2). Upon intracellular entry, ADCs transit to early endosomes and lysosomes, where they degrade under acidic conditions or with enzymatic activity. This breakdown releases cytotoxic drugs, which subsequently bind to DNA or microtubule proteins, inducing cell cycle arrest and eventual apoptosis [15]. The advancement of Trop-2-targeted ADCs has seen rapid progress in recent years, with multiple compounds currently undergoing clinical investigation [16,17,18,19].

3.1.1. Sacituzumab Govitecan

Sacituzumab govitecan (SG/IMMU-132) is the groundbreaking ADC that targets Trop-2. It comprises the humanized IgG1 monoclonal antibody hRS7 (KD = 0.3 nM) attached to SN-38, a topoisomerase inhibitor, via the cleavable linker CL2A. This innovative design targets patients with breast cancer and urothelial cancer [20,21,22]. The pH-sensitive CL2A linker facilitates the intracellular and extracellular release of the active payload, promoting a bystander effect that enhances the drug’s efficacy [23]. Moreover, SG has shown promising results in advanced and metastatic non-small-cell carcinoma (NSCLC) and invasive bladder cancer [24,25,26]. Its successful market introduction has set a precedent for Trop-2-targeted cancer therapy. The final study results of the phase III clinical trial TROPiCS-02, released in August 2023, demonstrated the efficacy of SG in patients with hormone receptor-positive/human epidermal growth factor receptor-2-negative metastatic breast cancer (HR+/HER2-mBC), leading to a significant improvement in their progression-free survival [27]. The data from TROPiCS-02 have resulted in the inclusion of SG as a category I option in the 2023 NCCN Guidelines for Breast Cancer and its recommendation as a second-line treatment for HR+/HER2− breast cancer patients who have previously received endocrine therapy (ET) and CDK4/6 inhibitors [21].

3.1.2. Datopotamab Deruxtecan

Datopotamab deruxtecan (Dato-DXd), the second ADC drug developed using the DXd technology platform, exhibits a potent bystander effect in the treatment of breast cancer and non-small-cell lung cancer [28]. MAAP-9001a, a humanized IgG1 antibody with low affinity (KD = 27 nM) to Trop-2 targets, was intended to broaden the drug’s therapeutic window, balancing efficacy and safety. The results from the TROPION-Lung01 study suggest that Dato-DXd monotherapy improves survival outcomes in locally advanced/metastatic NSCLC patients, regardless of actionable genomic alterations (AGA) status. Specifically, non-squamous cell carcinoma NSCLC patients demonstrate a particularly strong response to Dato-DXd treatment [29]. The phase II clinical trial TROPION-Lung05 further demonstrates the substantial anti-tumor activity and manageable safety profile of Dato-DXd in AGA patients with advanced/metastatic NSCLC who have undergone multiple lines of therapy [30,31]. Additionally, Dato-DXd displays promising efficacy and safety in the triple-negative breast cancer (TNBC) cohort and the HR+/HER2− cohort of TROPION-PanTumor01 [32,33]. Further clinical studies are underway to explore the potential application of Dato-DXd in HER2-negative breast cancer [34,35].

3.1.3. SKB264

SKB264 is an innovative ADC that targets Trop-2, utilizing the same antibody component as IMMU-132. The payload consists of a derivative of belotecan, a potent topoisomerase I inhibitor known as KL610023 (T030), linked via an enzymatically cleavable linker to enhance stability and reduce toxic side effects [36,37]. Initial clinical results show that SKB264 effectively treats advanced TNBC and other Trop-2-expressing solid tumors, with manageable side effects. Ongoing clinical trials are investigating the safety, tolerability, and objective response rate of SKB264 as a monotherapy or in combination with other treatments for advanced and metastatic NSCLC.

3.1.4. SHR-A1921

SHR-A1921 comprises a unique topoisomerase I inhibitor, SHR9265, connected to a proprietary IgG1 monoclonal antibody with a cleavable linker [19]. This ADC demonstrates superior binding affinity to human and cynomolgus monkey Trop-2 compared to existing ADCs targeting Trop-2, resulting in enhanced therapeutic effectiveness. The carefully designed spatial arrangement of the payload minimizes non-specific cleavage, ensuring sustained drug activity in vivo. Additionally, the high lipophilicity of the payload allows it to overcome tumor heterogeneity and resistance, producing a bystander effect. In clinical comparisons, SHR-A1921 exhibits a significantly longer half-life than SKB264, suggesting the potential for less frequent dosing and reduced patient burden. Overall, with its deep tissue penetration, optimized drug-to-antibody ratio, and proven stability and efficacy in preclinical trials, SHR-A1921 has emerged as a strong candidate in Trop-2-targeted ADC therapy.
As additional clinical trial data are gathered, it is anticipated that more Trop-2-targeted ADC medications will receive approval, offering novel treatment avenues for individuals battling cancer.

3.2. Drug Combination Therapy

The potential of ADCs in cancer treatment is hindered by the heterogeneity and individual variability of malignant tumors, making it difficult to achieve optimal therapeutic efficacy using a single agent or therapy. Consequently, the focus has shifted towards exploring combination therapies that involve the use of multiple drugs. This approach is seen as a promising strategy in anti-tumor drug research, as it allows for the targeting of various pathways and mechanisms of cancer cell growth and proliferation. Several clinical trials to investigate potential drug combination strategies are currently in progress (refer to Table 1 for details).

3.2.1. Combination with Chemotherapy

Combining ADCs with chemotherapeutic agents enhances the anti-tumor effect and reduces drug resistance. However, it is crucial to consider the timing of drug administration and potential drug toxicity when using combination regimens. Global clinical trials are currently in progress to assess the effectiveness of Dato-DXd in combination with platinum-based chemotherapy in different settings. For example, the TROPION-Lung02 study assessed the Trop-2-targeted ADC in combination with immunotherapy (pembrolizumab) ± platinum-based chemotherapy in advanced NSCLC. This study demonstrated that the combination of Dato-DXd with immune checkpoint inhibitors and chemotherapy was well-tolerated and had significant anti-tumor activity in patients with advanced NSCLC. In terms of safety, 61% of patients experienced grade ≥ 3 treatment-emergent adverse events (TEAEs), with common occurrences including nausea, anemia, malaise, and stomatitis [38]. Ongoing studies such as TROPION-Lung07 and AVANZAR are further exploring the potential of Trop-2-targeted ADC combination therapy. The TROPION-Lung07 study is explicitly investigating the efficacy and safety of Dato-DXd + immune checkpoint inhibitor ± chemotherapy in patients with advanced/metastatic NSCLC with PD-L1 < 50% [39]. On the other hand, the AVANZAR study will assess the efficacy and safety of Dato-DXd + dolvarizumab + carboplatin as a first-line treatment for advanced/metastatic NSCLC [40].

3.2.2. Combination with Molecular-Targeted Drugs

Molecular-targeted drugs encompass two main categories: those that target tumor cells directly and those that target the tumor growth microenvironment. The molecular-targeted drugs currently used in conjunction with Trop-2-targeted ADCs include PARP inhibitors, ATR inhibitors, and CDK4/6 inhibitors, among others. The combination of molecular-targeted drugs and ADCs shows significant promise in enhancing anti-tumor effects, reducing drug toxicity, and overcoming ADC resistance. The phase Ib SEASTA study demonstrated that the combination of rucaparib and SG exhibited potent anti-tumor activity in patients with advanced solid tumors, including those who had previously received PARP inhibitors and tumors with or without HRR gene mutations [41]. Another phase I study indicated that the ATR inhibitor berzosertib combined with SG showed a favorable safety profile compared to traditional chemotherapy without dose limitations. This combination also led to a lower occurrence of hematological toxicity than the combination of berzosertib and the conventional TOP1 inhibitor topotecan [42]. A phase II trial is currently underway to assess the safety and effectiveness of SG in combination with a CDK4/6 inhibitor (trilaciclib) in patients with TNBC. The initial data from the first 18 patients revealed a significant reduction in multiple associated adverse events compared to the results obtained using SG as a single agent [43]. Furthermore, the use of Ceralasertib (ATRi) in combination with Dato-DXd demonstrated the ability to re-sensitize Dato-DXD-resistant NCI-N87 cells in vitro and in vivo, potentially offering new strategies for combating ADC resistance and identifying biological markers of drug sensitivity [44].
Tumor progression requires the induction of tumor angiogenesis, with pro-angiogenic factors such as VEGF and PDGF mainly produced by tumor and stromal cells in the tumor microenvironment. Increased vascularization contributes to tumor recurrence, progression, invasion, and metastasis. The combination of antiangiogenic drugs with ADCs has the potential to target both tumor cells and the microenvironment, with current clinical trials exploring this approach in various cancer types. This strategic combination is expected to improve therapeutic outcomes, reduce the development of drug resistance, and potentially minimize side effects compared to monotherapy, offering promising options for cancer patients [45].

3.2.3. Combination with Immunotherapy

In clinical trials, immune checkpoint inhibitors (CPIs) such as CTLA-4 antibodies, PD-1 antibodies, and PD-L1 antibodies have been utilized in conjunction with Trop-2-targeted ADCs [46,47,48,49]. Research indicates that ADCs can stimulate the infiltration of CD8+ T-cells into tumor tissue, with these cells being the primary effector population driving anti-tumor responses in many cancer immunotherapy settings. ADCs promote anti-tumor immunity by activating and maturing dendritic cells through the release of cytotoxic payloads in the tumor microenvironment. This activation boosts the immune response to tumors. Additionally, certain cytotoxic compounds in ADCs can cause immunogenic cell death, leading to the release of ‘danger signals’, like calreticulin, HMGB1, and ATP, which trigger dendritic cell maturation and activation. These molecules are recognized by specific cell surface receptors on dendritic cells, further enhancing the immune response. The combination of these mechanisms of action of ADC payloads ultimately leads to the cross-priming of CD8+ T-cells in the lymph nodes by activated dendritic cells. This process promotes the infiltration of CD8+ T-cells into the tumor core, where they can engage in the cytolytic killing of tumor cells through the release of granzyme B and perforin by effector T-cells. Moreover, the abundance of CD8+ T-cells has been suggested as a means of predicting the efficacy of immune checkpoint-suppression therapy [50]. Essentially, ADCs have the potential to enhance anti-tumor effectiveness by prompting CD8+ T-cells to penetrate the tumor and heighten its responsiveness to CPI treatment [51]. A notable example is the TROPHY-U-01 study, a multi-cohort, open-label, phase II trial. The first cohort of this study involved metastatic urothelial carcinoma (mUC) patients who had previously experienced treatment failure with platinum chemotherapy and CPI [22]. According to an analysis of data from various cohorts, SG (SG) demonstrated encouraging effectiveness and a consistent positive outcome in mUC patients who were either new to platinum treatment or resistant to it. Furthermore, the combination of SG and CPI could offer a new therapeutic approach for mUC patients who have not been previously treated with CPI [22,47,52]. Additionally, the BEGONIA study was specifically designed to assess the effectiveness and safety of combining durvalumab with Dato-DXd as a first-line treatment for TNBC. The preliminary data from this study indicated a favorable safety profile and a noteworthy response rate with the combination therapy. An ongoing translational data analysis is being conducted to further substantiate the efficacy and safety of this treatment approach [53].

3.2.4. Combination with Other ADCs

The co-expression of multiple targets in the same cancer presents an opportunity to enhance treatment outcomes through the development of bi-specific ADCs or combination ADC therapies. For instance, in urothelial carcinoma, both NECTIN4 and Trop2/TACSTD2 are frequently overexpressed, and their expressions are closely linked. Additionally, HER2/ERBB2 is correlated with ERBB3 and Trop2/TACSTD2 [54]. The combination of ADCs targeting different receptors represents an innovative approach to cancer therapy. Synergistic effects can be achieved by concurrently targeting multiple crucial survival pathways in tumor cells to enhance treatment efficacy and mitigate the development of drug resistance. The initial results from a phase I trial combining two ADCs in the treatment of malignant tumors demonstrate that the combination of SG and EV (enfortumab vedotin) is both safe and feasible in patients with drug-resistant metastatic uroepithelial carcinoma, leading to a high early response rate [55]. Currently, a phase II clinical study (NCT06100874) is enrolling individuals to evaluate the safety and effectiveness of SG in combination with trastuzumab for metastatic HER2+ breast cancer.

3.2.5. Other Potential Combination Strategies

Radiation therapy is a frequently utilized method in the treatment of cancer, activating an immune response against tumors through the induction of cancer cell death and the release of antigens from within tumor cells. Precise levels of radiation have the ability to create new antigens and boost the immune response against tumors that have a high rate of mutation [56]. Furthermore, endocrine therapy, known for its low toxicity and good tolerability, particularly benefits hormone-dependent tumors like breast, prostate, and endometrial cancers. These theoretical foundations suggest that combining ADCs with radiotherapy or endocrine therapy could be promising for cancer patients. Two ongoing clinical trials are investigating the feasibility and safety of HER2-targeted ADCs combined with radiotherapy or endocrine therapy [57,58], offering new insights into the combined use of Trop-2-targeted ADCs. In patients with metastatic castration-resistant prostate cancer (mCRPC), the expression of TACSTD is positively correlated with AR-V7, indicating that Trop-2-targeted therapy in conjunction with endocrine therapy could potentially decrease hormone resistance [59]. A clinical trial of phase II is currently in progress to evaluate the efficacy and safety of SG in patients with mCRPC who are undergoing second-generation AR-targeted therapy (NCT03725761). More combination therapy strategies are being investigated, including the use of SG in combination with hydroxychloroquine for breast cancer (NCT06328387), and SG in combination with loperamide and G-CSF to enhance the tolerance of SG in patients with unresectable locally advanced or metastatic TNBC (NCT05520723).

4. Limitations

Despite advancements in Trop-2-targeted ADC therapies, challenges persist. These include the variability in Trop-2 expression, the stability of and resistance to ADCs, and the lack of predictive biomarkers of efficacy.

4.1. The variability in Trop-2 Expression

The expression pattern of Trop-2 in malignancies is complex. Its expression levels vary significantly between different types and stages of tumors and even within different regions of the same tumor. Toda et al. found that Trop-2 expression was significantly higher in anaplastic thyroid carcinoma (ATC) undifferentiated from papillary thyroid carcinoma (PTC) compared to ATC undifferentiated from follicular thyroid carcinoma (FTC) and de novo ATC. An immunohistochemistry (IHC) evaluation was conducted using the Allred scoring system, which involved grading specimens based on their intensity score (IS) and proportion score (PS). The IS classified specimens into four grades: 0 for negative, 1 for weak, 2 for moderate, and 3 for strong. PS determined the percentage of stained cells: 0 for 0%, 1 for <1%, 2 for 1–10%, 3 for 11–33%, 4 for 34–66%, and 5 for 67–100%. The total score (TS) was calculated by summing the IS and PS, with a rating of positive given when the TS was 3 or higher. The total score of Trop-2 was notably increased in the PTC origin-type group in comparison to the FTC origin-type and de novo-type groups [60]. Using RNA-seq data, Sperger et al. analyzed 634 metastatic castration-resistant prostate cancer (mCRPC) samples from four separate cohorts to compare TACSTD2 expression levels in tumors classified as adenocarcinoma or neuroendocrine prostate cancer (NEPC). Further sub-classification of patients with adenocarcinoma into luminal or basal subtypes revealed that tumors from these patients had markedly elevated TACSTD2 expression compared to tumors from patients with NEPC [61]. Studies on colorectal cancer have shown that moderately to highly differentiated tumor tissues exhibit increased Trop-2 expression, which is positively associated with CD9. The protein levels of Trop-2 and CD9 were examined through IHC in a case series involving 177 CRC patients. High levels of Trop-2 and CD9 were detected in moderately and poorly differentiated CRC tissues, while their expression was low in well-differentiated tissues. Furthermore, a biparametric analysis of Trop-2/CD9 in CRC identified subgroups where patients with high levels of both proteins had a significantly higher risk of metastatic relapse compared to those with low levels of Trop-2 and CD9 [62]. Moretto et al. noted significant differences in Trop-2 expression in metastatic colorectal cancer samples with varying proportions, from low to high expression, closely linked to BRAF gene mutation and tumor sites. In their study, a total of 251 tumor blocks and 135 paraffin slides from the TRIBE2 study were analyzed for Trop-2 immunohistochemical assessment. The results revealed a diverse expression pattern of Trop-2, with 23% classified as high, 30% as medium, and 47% as low. Tumors with high Trop-2 expression were more likely to have BRAF mutations and be located on the right side compared to those with medium and low expression levels [63]. Izci et al. confirmed the variability of Trop-2 expression levels across different disease stages and subtypes of TNBC. The study analyzed Trop-2 expression in TNBC patients diagnosed between 2000 and 2017 using IHC on whole-slide tumor sections. The expression levels were evaluated as continuous and categorical variables, with H-scores categorized as high (201–300), medium (100–200), and low (<100). The results showed that Trop-2 expression was high in 97 (16.5%) patients, medium in 149 (25.3%) patients, and low in 343 (58.2%) patients [64]. In oral squamous cell carcinoma (OSCC), Erber et al. identified a significant correlation between high central tumoral and lower peripheral Trop-2 expression patterns and impaired Recurrence-Free Survival (RFS). Their study, using IHC, examined Trop-2 expression in both central and peripheral tumor regions, as well as non-neoplastic oral mucosa. OSCC patients with high central and low peripheral Trop-2 expression had a five-year overall survival (OS) rate of 41.2%, compared to 55.6% for those with different expression patterns. The study revealed that high central tumoral and lower peripheral Trop-2 expression were associated with reduced OS and RFS in OSCC patients, even after adjusting for other factors in the multivariate Cox regression analysis [59]. In summary, the diverse expression patterns of Trop-2 present a considerable challenge for therapeutic strategies.

4.2. The Stability of and Resistance to ADCs

The stability of ADCs is crucial for their efficacy and safety. To effectively target tumor cells in vivo, ADC drugs must accurately and promptly deliver cytotoxic payloads. A detailed analysis revealed that the toxicity, dose-limiting effects, and maximum tolerated dose of ADC drugs were not directly related to the level of target antigen expression, but rather to the type of linker utilized [65]. Research has shown that drug instability can cause premature dissociation in vivo, leading to nonspecific release and subsequent adverse effects [66]. Despite efforts to improve the stability of ADC linkers, treatment-related adverse reactions, especially dose-limiting hematotoxicity, are common in clinical trials. For instance, the phase I/II IMMU-132-01 basket trial demonstrated IMMU-132’s efficacy in advanced TNBC but reported significant treatment-related adverse events, like nausea, diarrhea, fatigue, alopecia, and neutropenia [67]. Similarly, the ASCENT study, a phase III clinical trial for advanced TNBC treatment, revealed that patients receiving SG experienced more adverse events than those on traditional monotherapy, including neutropenia, leukopenia, diarrhea, anemia, and febrile neutropenia [16]. A fatal adverse event related to SG involving septic shock from neutropenic colitis was reported in TROPiCS-02 [27].
Additionally, mechanisms of resistance to ADCs, including altered target antigen expression, defective internalization pathways, and lysosomal dysfunction, have been a focus of research [68]. Recent studies have highlighted the production of tumor neoantigens in response to low-dose radiation as a potential strategy to address ADC resistance [56]. Nevertheless, the non-specific effects of an off-target payload remain a significant factor contributing to various tolerance phenomena observed in clinical settings [66].

4.3. Lack of Predictive Biomarkers of Efficacy

The levels of estrogen and progesterone receptors, along with human epidermal growth factor receptor 2 (HER-2), have become key biomarkers in breast cancer treatment for hormone therapy and targeted HER-2 therapy. Despite the potential value of Trop-2 expression levels in affecting sensitivity to Trop-2-targeted therapy, current treatment guidelines do not require Trop-2 expression levels to be tested before the use of Trop-2-targeted ADCs for breast or uroepithelial carcinoma patients. Clinical study analyses revealed that patients with varying Trop-2 expression levels derive benefits from ADC treatment, but the high expression does not significantly correlate with improved outcomes, possibly due to the bystander effect [69,70,71,72]. Other biomarkers associated with Trop-2 expression, such as SLFN11 (a predictor of SN-38 treatment sensitivity), Rad51 (involved in homologous recombination repair), and ERCC1, can also serve as important predictors of treatment outcomes [73].

5. Future Direction

5.1. Antibody Design

Further research into the function of Trop-2 in normal and tumor cells will enhance our understanding of Trop-2’s potential and limitations as a therapeutic target. This will enable the development of more precise antibodies. Trop-2 cleavage by ADAM10 is prevalent in various tumor types, such as skin, ovarian, colon, and breast cancer, while not being observed in normal cells [74,75]. The cancer-specific antibody Hu2G10, developed based on this finding, exhibits a significantly higher affinity for cancer-specific Trop-2 compared to wtTrop-2. In vivo studies demonstrate that the naked Hu2G10 antibody effectively inhibits the growth of breast cancer, colon cancer, ovarian cancer, and prostate cancer, thereby enhancing anti-cancer efficacy and paving the way for the next generation of ADCs [76,77,78,79]. Furthermore, exploring the heterogeneity of Trop-2 is expected to lead to the development of antibodies with improved affinity. Trop-2 molecules have been found to form stable dimers and polymers at interfaces of cancer cells, potentially hindering access to antibody-binding sites in densely packed cancer cell populations. The 2EF mAb was specifically designed to overcome this challenge and has shown enhanced efficacy against various tumor models, penetrating deeper into cancer sites, while also exhibiting reduced toxicity in animal studies. Consequently, the humanized antibody Hu2EF-7 has emerged as a promising candidate for the development of novel anti-Trop-2 ADCs [80].

5.2. Influence of Extracellular Vesicles on ADCs

Extracellular vesicles (EVs) are small membrane structures secreted by cells that play a crucial role in maintaining cellular physiological balance and in pathological processes. They are categorized into exosomes and microvesicles and are capable of transporting proteins, lipids, RNA, and other biological molecules into the extracellular environment [81]. The exceptional target specificity, biocompatibility, low immunogenicity, and inherent stability of extracellular vesicles have positioned them as promising drug delivery systems in recent years, with significant potential in precision tumor therapy. Furthermore, extracellular vesicles released by tumor cells may carry specific biomarkers, offering a novel approach for early tumor diagnosis, prognosis assessment, and therapeutic monitoring. Moreover, these vesicles can modulate communication among tumor cells by promoting angiogenesis and modifying the extracellular matrix, thus impacting tumor growth and metastasis [82,83]. The study of extracellular vesicles in tumor therapy is currently focused on their potential and practicality as drug delivery vehicles.
Extracellular vesicles can improve drug targeting and stability [84,85,86]. Yong et al. provided theoretical validation for exosomes as effective targeted drug carriers for cancer therapy [87]. Li et al. discovered that drug-loaded extracellular vesicles with specific targeting ligands coated with high-density antibodies can enhance their ability to target tumors and effectively combat cancer [88]. Saari et al. utilized tumor cell-derived EVs to deliver the drug PTX, showcasing its enhanced targeting and cytotoxicity against prostate cancer [89]. Niu et al. created bionic EV-DNs that can surpass the limitations of the blood–brain tumor barrier, enabling efficient drug delivery and significantly enhancing anti-tumor efficacy [90], while Zhang et al. demonstrated that liposomes can effectively deliver drugs to the brain without compromising the integrity of the blood–brain barrier [91]. Feng et al. showed that folic acid assembly in extracellular vesicles can improve drug delivery, regulate the tumor microenvironment, and directly inhibit tumor metastasis, thus enhancing therapeutic outcomes [92].
Extracellular vesicles (EVs) expressing Trop-2 have the potential to enhance the effectiveness of Trop-2-targeted ADCs by delivering drugs to Trop-2-negative tumor cells or tumor stromal cells. For instance, brentuximab vedotin and trastuzumab emtansine can utilize EVs carrying CD30-positive and HER2-positive markers released by tumor cells to target neighboring tumor cells lacking these antigens, leading to tumor regression [93,94,95] (Figure 1). Prostate cancer cells release extracellular vesicles (EVs) that express Trop-2 [96]. These EVs have the potential to transfer their contents to cancer cells in the same tumor that do not express Trop-2, causing a bystander effect. Additionally, Trop-2-expressing EVs could potentially transport ADCs to other cells in the tumor microenvironment, thereby inhibiting cancer proliferation [97].
Extracellular vesicles can also influence drug resistance. Wang et al. demonstrated that extracellular vesicles derived from human gastric epithelial cells can deliver miR-214 to overcome cisplatin chemotherapy resistance in gastric cancer [98]. Liang et al. utilized engineered extracellular vesicles to deliver anti-tumor drugs (5-FU and miR-21 inhibitors), resulting in enhanced tumor cytotoxicity and the reversal of cancer cell resistance to 5-FU [99].
In conclusion, EVs offer new strategies for further optimizing ADC design. The high biocompatibility and low immunogenicity of EVs can effectively improve the distribution and release efficiency of ADCs. As promising drug delivery vehicles, EVs have great potential in improving efficacy, reducing toxicity, and overcoming drug resistance to Trop-2-targeted ADCs.

5.3. Multi-Target Combination Therapy

The co-expression of multiple different ADC targets within the same tumor provides an opportunity for the development of future clinical trials involving combinations of ADCs or the creation of bi-specific ADCs, for example, combining anti-NECTIN4 with anti-Trop-2 in lung, breast, and ovarian cancer or anti-HER2 with anti-ERBB3 in lung, breast, and prostate cancer [54,63]. Additionally, in solid tumors with high Trop-2 expression, various other potential therapeutic targets exhibit heterogeneity in specific tumor types. For example, there is an increase in FOXM1 expression in breast cancer, hepatocellular carcinoma, and basal cell carcinoma. TP63 and WT1 are also prominently expressed in squamous cell carcinoma, bladder cancer, breast cancer, and gastric cancer [100,101,102]. In colorectal cancer, the positive correlation between Trop-2 and CD9 expression opens up opportunities for combined targeted therapy strategies [62].
One promising future direction for ADCs is to investigate combination strategies of different target ADCs to improve drug efficacy. Optimizing combination therapy through rational administration sequences and dose combinations is essential. Furthermore, exploring ADCs with dual targets or multiple payloads is a worthwhile consideration.

5.4. Personalized Treatment

Tools to predict the drug sensitivity of each patient can be helpful in guiding treatment decisions. At present, there is no ideal strategy to assist clinicians in choosing the appropriate treatment for patients. While traditional tumor classification methods can help guide treatment to a certain extent, they cannot effectively identify the optimal group for Trop-2-targeted medication. The effectiveness of ADCs is closely related to their mechanism of action. The accurate identification and selection of potential responsive populations have emerged as pressing issues in the field of tumor therapy. Most of the currently approved Trop-2-targeted ADCs are inhibitors of topoisomerase 1 (TOP1), making high TOP1 expression a potential indicator of sensitivity to camptothecin analogues. Furthermore, the presence of the SN-38-sensitivity predictor SLFN11, HRR-related protein Rad51, and ERCC1 has shown promise in predicting clinical benefits [73,103]. Additionally, CTSL, which hydrolyzes T-DXd peptide ligators, is elevated in advanced breast cancer. BCRP/ABCG2, responsible for mediating camptothecin’s effects and contributing to the bystander effect observed with T-DXd, is upregulated in metastatic compared to primary breast cancer, among other types [54]. Although this field of research is still in its early stages, it is a crucial step towards achieving precision medicine. Therefore, the precise identification of populations sensitive to ADC treatment is a key direction for future exploration. Table 2 displays the relevant clinical trials.

6. Discussion

Precision targeted therapy, particularly in the individualized management of complex conditions like cancer, holds significant therapeutic promise. Future progress relies on advancing novel technologies, wider biomarker utilization, and a deeper understanding of underlying causes. Peptide–drug conjugates (PDCs) are the upcoming wave of targeted treatments after ADCs, providing better cell penetration and increased drug specificity [104]. PDC is a targeted therapeutic agent with a mechanism of action similar to that of ADCs, involving the conjugation of peptides and drugs to facilitate the effective delivery of chemotherapy agents to tumor cells. The three essential components of PDC include homing peptides, linkers, and cytotoxic drugs, which synergistically target receptors on tumor cells and enhance the therapeutic efficacy of peptide treatments. Currently, pharmaceutical firms are actively creating PDCs as specialized therapeutic options for ailments like cancer and metabolic disorders. PDCs demonstrate effective tumor infiltration, economical production, and a lack of immunogenicity, making them more versatile in the commercial and clinical sectors compared to other merged drugs. However, ADCs possess unique benefits over PDCs. ADCs can precisely bind to target cell surface antigens due to high affinity, enabling accurate drug delivery. They also induce strong cell killing effects through antibody-mediated cytotoxicity and complement-dependent cytotoxicity mechanisms. Furthermore, ADC technology allows toxic compounds to be stably linked to antibodies, increasing local drug concentration without systemic toxicity, enhancing therapeutic outcomes. As a whole, ADCs and PDCs are pivotal in contemporary medicine, enhancing treatment personalization and effectiveness through insights from molecular biology.
Trop-2-targeted ADCs have demonstrated substantial promise in the management of diverse solid malignancies. In the future, we look forward to improving the efficacy of Trop-2-targeted therapies by optimizing antibody design and exploring new combination therapy strategies and more personalized treatment options for cancer patients. Subsequent research is necessary to investigate how EVs can be engineered or modified to enhance their specificity and efficiency in delivering ADCs, as well as to identify more efficient biological markers of drug sensitivity.

Author Contributions

Conceptualization, Y.J., J.L. (Jiahao Li) and X.X.; writing—original draft preparation, Y.J., H.Z., J.L. (Junxia Liu) and W.H.; writing—review and editing, Y.J.; supervision, T.C. and H.X. All authors have read and agreed to the published version of the manuscript.

Funding

The authors gratefully acknowledge the financial support from Beijing Xisike Clinical Oncology Research Foundation (Y-HR2020MS-1039).

Conflicts of Interest

The authors declare no competing interests.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Ismael, G.F.; Rosa, D.D.; Mano, M.S.; Awada, A. Novel cytotoxic drugs: Old challenges, new solutions. Cancer Treat. Rev. 2008, 34, 81–91. [Google Scholar] [CrossRef] [PubMed]
  3. Boakes, R.A.; Tarrier, N.; Barnes, B.W.; Tattersall, M.H. Prevalence of anticipatory nausea and other side-effects in cancer patients receiving chemotherapy. Eur. J. Cancer 1993, 29, 866–870. [Google Scholar] [CrossRef] [PubMed]
  4. Green, M.R. Targeting Targeted Therapy. N. Engl. J. Med. 2004, 350, 2191–2193. [Google Scholar] [CrossRef]
  5. Birrer, M.J.; Moore, K.N.; Betella, I.; Bates, R.C. Antibody-Drug Conjugate-Based Therapeutics: State of the Science. JNCI J. Natl. Cancer Inst. 2019, 111, 538–549. [Google Scholar] [CrossRef]
  6. Li, W.Q.; Guo, H.-F.; Li, L.-Y.; Zhang, Y.-F.; Cui, J.-W. The promising role of antibody drug conjugate in cancer therapy: Combining targeting ability with cytotoxicity effectively. Cancer Med. 2021, 10, 4677–4696. [Google Scholar] [CrossRef]
  7. Drago, J.Z.; Modi, S.; Chandarlapaty, S. Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat. Rev. Clin. Oncol. 2021, 18, 327–344. [Google Scholar] [CrossRef] [PubMed]
  8. Guerra, E.; Trerotola, M.; Alberti, S. Targeting Trop-2 as a Cancer Driver. J. Clin. Oncol. 2023, 41, 4688–4692. [Google Scholar] [CrossRef]
  9. Syed, Y.Y. Sacituzumab Govitecan: First Approval. Drugs 2020, 80, 1019–1025. [Google Scholar] [CrossRef]
  10. Fornaro, M.; Dell’ Arciprete, R.; Stella, M.; Bucci, C.; Nutini, M.; Capri, M.G.; Alberti, S. Cloning of the gene encoding Trop-2, a cell-surface glycoprotein expressed by human carcinomas. Int. J. Cancer 1995, 62, 610–618. [Google Scholar] [CrossRef]
  11. Calabrese, G.; Crescenzi, C.; Morizio, E.; Palka, G.; Guerra, E.; Alberti, S. Assignment of TACSTD1 (alias TROP1, M4S1) to human chromosome 2p21 and refinement of mapping of TACSTD2 (alias TROP2, M1S1) to human chromosome 1p32 by in situ hybridization. Cytogenet. Cell Genet. 2001, 92, 164–165. [Google Scholar] [CrossRef]
  12. Vidmar, T.; Pavšič, M.; Lenarčič, B. Biochemical and preliminary X-ray characterization of the tumor-associated calcium signal transducer 2 (Trop2) ectodomain. Protein Expr. Purif. 2013, 91, 69–76. [Google Scholar] [CrossRef]
  13. Goldenberg, D.M.; Stein, R.; Sharkey, R.M. The emergence of trophoblast cell-surface antigen 2 (TROP-2) as a novel cancer target. Oncotarget 2018, 9, 28989–29006. [Google Scholar] [CrossRef]
  14. Lenárt, S.; Lenárt, P.; Šmarda, J.; Remšík, J.; Souček, K.; Beneš, P. Trop2: Jack of All Trades, Master of None. Cancers 2020, 12, 3328. [Google Scholar] [CrossRef]
  15. Tsuchikama, K.; An, Z. Antibody-drug conjugates: Recent advances in conjugation and linker chemistries. Protein Cell 2018, 9, 33–46. [Google Scholar] [CrossRef]
  16. 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]
  17. Okajima, D.; Yasuda, S.; Maejima, T.; Karibe, T.; Sakurai, K.; Aida, T.; Toki, T.; Yamaguchi, J.; Kitamura, M.; Kamei, R.; et al. Datopotamab Deruxtecan, a Novel TROP2-directed Antibody–drug Conjugate, Demonstrates Potent Antitumor Activity by Efficient Drug Delivery to Tumor Cells. Mol. Cancer Ther. 2021, 20, 2329–2340. [Google Scholar] [CrossRef]
  18. Fang, W.; Cheng, Y.; Chen, Z.; Wang, W.; Yin, Y.; Li, Y.; Xu, H.; Li, X.; Wainberg, Z.A.; Yu, G.; et al. SKB264 (TROP2-ADC) for the treatment of patients with advanced NSCLC: Efficacy and safety data from a phase 2 study. J. Clin. Oncol. 2023, 41, 9114. [Google Scholar] [CrossRef]
  19. He, N.; Yang, C.; Yang, Y.; Xue, Z.; Xu, J.; Zhao, L.; Feng, J.; Ye, X.; Zhang, Z.; He, F. Abstract LB030: SHR-A1921, a novel TROP-2 ADC with an optimized design and well-balanced profile between efficacy and safety. Cancer Res. 2023, 83 (Suppl. S8), LB030. [Google Scholar] [CrossRef]
  20. Cardillo, T.M.; Govindan, S.V.; Sharkey, R.M.; Trisal, P.; Arrojo, R.; Liu, D.; Rossi, E.A.; Chang, C.H.; Goldenberg, D.M. Sacituzumab Govitecan (IMMU-132), an Anti-Trop-2/SN-38 Antibody–Drug Conjugate: Characterization and Efficacy in Pancreatic, Gastric, and Other Cancers. Bioconjug. Chem. 2015, 26, 919–931. [Google Scholar] [CrossRef]
  21. Gradishar, W.J.; Moran, M.S.; Abraham, J.; Abramson, V.; Aft, R.; Agnese, D.; Allison, K.H.; Anderson, B.; Burstein, H.J.; Chew, H.; et al. NCCN Guidelines® Insights: Breast Cancer, Version 4.2023: Featured Updates to the NCCN Guidelines. J. Natl. Compr. Cancer Netw. 2023, 21, 594–608. [Google Scholar] [CrossRef]
  22. Tagawa, S.T.; Balar, A.V.; Petrylak, D.P.; Kalebasty, A.R.; Loriot, Y.; Fléchon, A.; Jain, R.K.; Agarwal, N.; Bupathi, M.; Barthelemy, P.; et al. TROPHY-U-01: A Phase II Open-Label Study of Sacituzumab Govitecan in Patients With Metastatic Urothelial Carcinoma Progressing After Platinum-Based Chemotherapy and Checkpoint Inhibitors. J. Clin. Oncol. 2021, 39, 2474–2485. [Google Scholar] [CrossRef] [PubMed]
  23. Goldenberg, D.M.; Cardillo, T.M.; Govindan, S.V.; Rossi, E.A.; Sharkey, R.M. Trop-2 is a novel target for solid cancer therapy with sacituzumab govitecan (IMMU-132), an antibody-drug conjugate (ADC). Oncotarget 2015, 6, 22496–22512. [Google Scholar] [CrossRef] [PubMed]
  24. Saxena, A.; Michel, L.S.; Hong, Q.; Hilsinger, K.; Kanwal, C.; Pichardo, C.; Goswami, T.; Santin, A. TROPiCS–03: A phase II open-label study of sacituzumab govitecan (SG) in patients with metastatic solid tumors. J. Clin. Oncol. 2020, 38, TPS3648. [Google Scholar] [CrossRef]
  25. Michel, L.; Jimeno, A.; Sukari, A.; Beck, T.; Chiu, J.; Ahern, E.; Hilton, J.; Hollebecque, A.; Zanetta, S.; Patel, J.; et al. 859MO Sacituzumab govitecan (SG) in patients (pts) with relapsed/refractory (R/R) advanced head and neck squamous cell carcinoma (HNSCC): Results from the phase II TROPiCS-03 basket trial. Ann. Oncol. 2023, 34, S558. [Google Scholar] [CrossRef]
  26. Dowlati, A.; Cervantes, A.; Babu, S.; Hamilton, E.; Wong, S.; Tazbirkova, A.; Sullivan, I.; de Lummen, C.V.M.; Italiano, A.; Patel, J.; et al. 1990MO Sacituzumab govitecan (SG) as second-line (2L) treatment for extensive stage small cell lung cancer (ES-SCLC): Preliminary results from the phase II TROPiCS-03 basket trial. Ann. Oncol. 2023, 34, S1061–S1062. [Google Scholar] [CrossRef]
  27. Rugo, H.S.; Bardia, A.; Marmé, F.; Cortés, J.; Schmid, P.; Loirat, D.; Trédan, O.; Ciruelos, E.; Dalenc, F.; Pardo, P.G.; et al. Overall survival with sacituzumab govitecan in hormone receptor-positive and human epidermal growth factor receptor 2-negative metastatic breast cancer (TROPiCS-02): A randomised, open-label, multicentre, phase 3 trial. Lancet 2023, 402, 1423–1433. [Google Scholar] [CrossRef]
  28. Ogitani, Y.; Hagihara, K.; Oitate, M.; Naito, H.; Agatsuma, T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody–drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 2016, 107, 1039–1046. [Google Scholar] [CrossRef]
  29. Ahn, M.J.; Lisberg, A.; Paz-Ares, L.; Cornelissen, R.; Girard, N.; Pons-Tostivint, E.; Baz, D.V.; Sugawara, S.; Dols, M.C.; Pérol, M.; et al. 509MO Datopotamab deruxtecan (Dato-DXd) vs docetaxel in previously treated advanced/metastatic (adv/met) non-small cell lung cancer (NSCLC): Results of the randomized phase III study TROPION-Lung01. Ann. Oncol. 2023, 34, S1665–S1666. [Google Scholar] [CrossRef]
  30. Kitazono, S.; Paz-Ares, L.; Ahn, M.J.; Lisberg, A.E.; Cho, B.C.; Blumenschein, G.; Shum, E.; Pons-Tostivint, E.; Goto, Y.; Yoh, K.; et al. 518MO TROPION-Lung05: Datopotamab deruxtecan (Dato-DXd) in previously treated non-small cell lung cancer (NSCLC) with actionable genomic alterations (AGAs). Ann. Oncol. 2023, 34, S1671–S1672. [Google Scholar] [CrossRef]
  31. Ahn, M.J.; Cho, B.; Goto, Y.; Yoh, K.; Su, W.-C.; Shimizu, J.; Lee, J.-S.; Oizumi, S.; Yang, J.-H.; Hayashi, H.; et al. 552P TROPION-Lung05: Datopotamab deruxtecan (Dato-DXd) in Asian patients (pts) with previously treated non-small cell lung cancer (NSCLC) with actionable genomic alterations (AGAs). Ann. Oncol. 2023, 34, S1684–S1685. [Google Scholar] [CrossRef]
  32. Bardia, A.; Krop, I.; Meric-Bernstam, F.; Tolcher, A.W.; Mukohara, T.; Lisberg, A.; Shimizu, T.; Hamilton, E.; Spira, A.I.; Papadopoulos, K.P.; et al. Abstract P6-10-03: Datopotamab Deruxtecan (Dato-DXd) in Advanced Triple-Negative Breast Cancer (TNBC): Updated Results From the Phase 1 TROPION-PanTumor01 Study. Cancer Res. 2023, 83 (Suppl. S5), P6-10-03. [Google Scholar] [CrossRef]
  33. Meric-Bernstam, F.; Krop, I.; Juric, D.; Kogawa, T.; Hamilton, E.; Spira, A.I.; Mukohara, T.; Tsunoda, T.; Damodaran, S.; Greenberg, J.; et al. Abstract PD13-08: PD13-08 Phase 1 TROPION-PanTumor01 Study Evaluating Datopotamab Deruxtecan (Dato-DXd) in Unresectable or Metastatic Hormone Receptor–Positive/HER2–Negative Breast Cancer (BC). Cancer Res. 2023, 83 (Suppl. S5), PD13-08. [Google Scholar] [CrossRef]
  34. Bardia, A.; Jhaveri, K.; Im, S.-A.; Simon, S.P.; De Laurentiis, M.; Wang, S.; Martinez, N.; Borges, G.S.; Cescon, D.; Hattori, M.; et al. LBA11 Datopotamab deruxtecan (Dato-DXd) vs chemotherapy in previously-treated inoperable or metastatic hormone receptor-positive, HER2-negative (HR+/HER2–) breast cancer (BC): Primary results from the randomised phase III TROPION-Breast01 trial. Ann. Oncol. 2023, 34, S1264–S1265. [Google Scholar] [CrossRef]
  35. Dent, R.; Cescon, D.W.; Bachelot, T.; Jung, K.H.; Shao, Z.-M.; Saji, S.; Traina, T.A.; Vuković, P.; Mapiye, D.; Maxwell, M.; et al. Abstract OT1-03-05: TROPION-Breast02: Phase 3, open-label, randomized study of first-line datopotamab deruxtecan versus chemotherapy in patients with locally recurrent inoperable or metastatic TNBC who are not candidates for anti-PD-(L)1 therapy. Cancer Res. 2023, 83 (Suppl. S5), OT1-03-05. [Google Scholar] [CrossRef]
  36. Liu, Y.; Lian, W.; Zhao, X.; Diao, Y.; Xu, J.; Xiao, L.; Qing, Y.; Xue, T.; Wang, J. SKB264 ADC: A first-in-human study of SKB264 in patients with locally advanced unresectable/metastatic solid tumors who are refractory to available standard therapies. J. Clin. Oncol. 2020, 38, TPS3659. [Google Scholar] [CrossRef]
  37. Cheng, Y.; Yuan, X.; Tian, Q.; Huang, X.; Chen, Y.; Pu, Y.; Long, H.; Xu, M.; Ji, Y.; Xie, J.; et al. Preclinical profiles of SKB264, a novel anti-TROP2 antibody conjugated to topoisomerase inhibitor, demonstrated promising antitumor efficacy compared to IMMU-132. Front. Oncol. 2022, 12, 951589. [Google Scholar] [CrossRef] [PubMed]
  38. Goto, Y.; Su, W.C.; Levy, B.P.; Rixe, O.; Yang, T.Y.; Tolcher, A.W.; Lou, Y.; Zenke, Y.; Savvides, P.; Felip, E.; et al. TROPION-Lung02: Datopotamab deruxtecan (Dato-DXd) plus pembrolizumab (pembro) with or without platinum chemotherapy (Pt-CT) in advanced non-small cell lung cancer (aNSCLC). J. Clin. Oncol. 2023, 41 (Suppl. S16), 9004. [Google Scholar] [CrossRef]
  39. Okamoto, I.; Kuyama, S.; Girard, N.; Lu, S.; Franke, F.; Pan, E.; Ren, N.; Chen, A.; Oputa, E.; Lisberg, A. 1505TiP TROPION-Lung07: A phase III trial of datopotamab deruxtecan (Dato-DXd) plus pembrolizumab (pembro) with or without platinum chemotherapy (Pt-CT) as first-line (1L) therapy in advanced/metastatic (adv/met) non-small cell lung cancer (NSCLC) with PD-L1 expression. Ann. Oncol. 2023, 34, S847–S848. [Google Scholar]
  40. Aggarwal, C.; Cheema, P.; Arrieta, O.; Bonanno, L.; Borghaei, H.; Hu, Y.; Kim, D.-W.; Junior, G.d.C.; Hepner, A.; Deng, C.; et al. P2.04-02 AVANZAR: Phase III Study of Datopotamab Deruxtecan (Dato-DXd) + Durvalumab + Carboplatin as 1L Treatment of Advanced/mNSCLC. J. Thorac. Oncol. 2023, 18, S305–S306. [Google Scholar] [CrossRef]
  41. Yap, T.; Hamilton, E.; Bauer, T.; Dumbrava, E.I.; Jeselsohn, R.; Enke, A.; Hurley, S.; Lin, K.; Wride, K.; Giordano, H.; et al. 547P Rucaparib+ sacituzumab govitecan (SG): Initial data from the phase Ib/II SEASTAR study (NCT03992131). Ann. Oncol. 2020, 31, S476–S477. [Google Scholar] [CrossRef]
  42. Abel, M.L.; Takahashi, N.; Peer, C.; Redon, C.E.; Nichols, S.; Vilimas, R.; Lee, M.-J.; Lee, S.; Shelat, M.; Kattappuram, R.; et al. Targeting Replication Stress and Chemotherapy Resistance with a Combination of Sacituzumab Govitecan and Berzosertib: A Phase I Clinical Trial. Clin. Cancer Res. 2023, 29, 3603–3611. [Google Scholar] [CrossRef] [PubMed]
  43. Pelosci, A. Trilaciclib Prior to Sacituzumab Govitecan-hziy Appeared to Reduce AEs in Advanced TNBC; Cancer Network, 2022. Available online: https://www.cancernetwork.com/view/trilaciclib-prior-to-sacituzumab-govitecan-hziy-appeared-to-reduce-aes-in-advanced-tnbc (accessed on 20 April 2024).
  44. Przybyla, A.; Rane, C.; Patel, G.; Bhavsar, D.; Wen, S.; Clark, D.; Kim, Y.J.; Lau, A.; Wallez, Y.; Mettetal, J.; et al. Abstract LB227: Leveraging novel Dato-DXd resistance models to inform biomarker discovery and rational combinations to combat drug resistance. Cancer Res. 2023, 83 (Suppl. S8), LB227. [Google Scholar] [CrossRef]
  45. Qiu, S.; Zhang, J.; Wang, Z.; Lan, H.; Hou, J.; Zhang, N.; Wang, X.; Lu, H. Targeting Trop-2 in cancer: Recent research progress and clinical application. Biochim. Biophys. Acta (BBA) Rev. Cancer 2023, 1878, 188902. [Google Scholar] [CrossRef]
  46. Jain, R.; Chahoud, J.; Chatwal, M.; Kim, Y.; Dhillon, J.; Vosoughi, A.; Araujo, C.; Li, R.; Zhang, J.; Sonpavde, G. 720TiP Phase I/II study of ipilimumab plus nivolumab (IPI-NIVO) combined with sacituzumab govitecan in patients with metastatic cisplatin-ineligible urothelial carcinoma. Ann. Oncol. 2021, 32, S724. [Google Scholar] [CrossRef]
  47. Grivas, P.; Pouessel, D.; Park, C.H.; Barthelemy, P.; Bupathi, M.; Petrylak, D.P.; Agarwal, N.; Gupta, S.; Flechon, A.; Ramamurthy, C.; et al. Primary analysis of TROPHY-U-01 cohort 3, a phase 2 study of sacituzumab govitecan (SG) in combination with pembrolizumab (Pembro) in patients (pts) with metastatic urothelial cancer (mUC) that progressed after platinum (PT)-based therapy. J. Clin. Oncol. 2023, 41, 518. [Google Scholar] [CrossRef]
  48. Tagawa, S.T.; Grivas, P.; Petrylak, D.P.; Sternberg, C.N.; Swami, U.; Bhatia, A.; Pichardo, C.; Goswami, T.; Loriot, Y. TROPHY-U-01 cohort 4: Sacituzumab govitecan (SG) in combination with cisplatin (Cis) in platinum (PLT)-naïve patients (pts) with metastatic urothelial cancer (mUC). J. Clin. Oncol. 2022, 40, TPS581. [Google Scholar] [CrossRef]
  49. Duran, I.; Necchi, A.; Powles, T.; Loriot, Y.; Ramamurthy, C.; Recio-Boiles, A.; Sweis, R.F.; Bedke, J.; Tonelli, J.; Sierecki, M.; et al. TROPHY-U-01 cohort 6: Sacituzumab govitecan (SG), SG plus zimberelimab (ZIM), SG plus ZIM plus domvanalimab (DOM), or carboplatin (CARBO)+ gemcitabine (GEM) in cisplatin-ineligible patients (pts) with treatment-naive metastatic urothelial cancer (mUC). J. Clin. Oncol. 2023, 41, TPS59. [Google Scholar] [CrossRef]
  50. Gerber, H.P.; Sapra, P.; Loganzo, F.; May, C. Combining antibody–drug conjugates and immune-mediated cancer therapy: What to expect? Biochem. Pharmacol. 2016, 102, 1–6. [Google Scholar] [CrossRef]
  51. Iwata, T.N.; Ishii, C.; Ishida, S.; Ogitani, Y.; Wada, T.; Agatsuma, T. A HER2-Targeting Antibody–Drug Conjugate, Trastuzumab Deruxtecan (DS-8201a), Enhances Antitumor Immunity in a Mouse Model. Mol. Cancer Ther. 2018, 17, 1494–1503. [Google Scholar] [CrossRef]
  52. Petrylak, D.P.; Tagawa, S.T.; Jain, R.K.; Bupathi, M.; Balar, A.V.; Rezazadeh, A.; George, S.; Palmbos, P.L.; Nordquist, L.T.; Davis, N.B.; et al. Primary analysis of TROPHY-U-01 cohort 2, a phase 2 study of sacituzumab govitecan (SG) in platinum (PT)-ineligible patients (pts) with metastatic urothelial cancer (mUC) that progressed after prior checkpoint inhibitor (CPI) therapy. J. Clin. Oncol. 2023, 41, 520. [Google Scholar] [CrossRef]
  53. Schmid, P.; Wysocki, P.; Ma, C.; Park, Y.; Fernandes, R.; Lord, S.; Baird, R.; Prady, C.; Jung, K.; Asselah, J.; et al. 379MO Datopotamab deruxtecan (Dato-DXd) + durvalumab (D) as first-line (1L) treatment for unresectable locally advanced/metastatic triple-negative breast cancer (a/mTNBC): Updated results from BEGONIA, a phase Ib/II study. Ann. Oncol. 2023, 34, S337. [Google Scholar] [CrossRef]
  54. Bosi, C.; Bartha, Á.; Galbardi, B.; Notini, G.; Naldini, M.M.; Licata, L.; Viale, G.; Mariani, M.; Pistilli, B.; Ali, H.R.; et al. Pan-cancer analysis of antibody-drug conjugate targets and putative predictors of treatment response. Eur. J. Cancer 2023, 195, 113379. [Google Scholar] [CrossRef]
  55. McGregor, B.A.; Sonpavde, G.; Kwak, L.; Regan, M.; Gao, X.; Hvidsten, H.; Mantia, C.; Wei, X.; Berchuck, J.; Berg, S.; et al. The Double Antibody Drug Conjugate (DAD) phase I trial: Sacituzumab govitecan plus enfortumab vedotin for metastatic urothelial carcinoma. Ann. Oncol. 2024, 35, 91–97. [Google Scholar] [CrossRef]
  56. Lussier, D.M.; Alspach, E.; Ward, J.P.; Miceli, A.P.; Runci, D.; White, J.M.; Mpoy, C.; Arthur, C.D.; Kohlmiller, H.N.; Jacks, T.; et al. Radiation-induced neoantigens broaden the immunotherapeutic window of cancers with low mutational loads. Proc. Natl. Acad. Sci. USA 2021, 118, e2102611118. [Google Scholar] [CrossRef] [PubMed]
  57. Xu, M.; Kong, Y.; Xing, P.; Chen, R.; Ma, Y.; Shan, C.; LiYuan, Z. A Multicenter, Single-Arm, Phase II Trial of RC48-ADC Combined with Radiotherapy, PD-1/PD-L1 Inhibitor Sequential GM-CSF and IL-2 (PRaG3. 0 regimen) for the Treatment of HER2-Expressing Advanced Solid Tumors. Int. J. Radiat. Oncol. Biol. Phys. 2022, 114, e428. [Google Scholar] [CrossRef]
  58. Fischer, L. Neoadjuvant Trastuzumab Deruxtecan ± Endocrine Therapy Demonstrates Promise in HR+ HER2-Low Breast Cancer; Cancer Network, 2022. Available online: https://www.cancernetwork.com/view/neoadjuvant-trastuzumab-deruxtecan-endocrine-therapy-demonstrates-promise-in-hr-her2-low-breast-cancer (accessed on 20 April 2024).
  59. Erber, R.; Spoerl, S.; Mamilos, A.; Krupar, R.; Hartmann, A.; Ruebner, M.; Taxis, J.; Wittenberg, M.; Reichert, T.E.; Spanier, G.; et al. Impact of Spatially Heterogeneous Trop-2 Expression on Prognosis in Oral Squamous Cell Carcinoma. Int. J. Mol. Sci. 2021, 23, 87. [Google Scholar] [CrossRef]
  60. Toda, S.; Sato, S.; Saito, N.; Sekihara, K.; Matsui, A.; Murayama, D.; Nakayama, H.; Suganuma, N.; Okubo, Y.; Hayashi, H.; et al. TROP-2, Nectin-4, GPNMB, and B7-H3 Are Potentially Therapeutic Targets for Anaplastic Thyroid Carcinoma. Cancers 2022, 14, 579. [Google Scholar] [CrossRef] [PubMed]
  61. Sperger, J.M.; Helzer, K.T.; Stahlfeld, C.N.; Jiang, D.; Singh, A.; Kaufmann, K.R.; Niles, D.J.; Heninger, E.; Rydzewski, N.R.; Wang, L.; et al. Expression and Therapeutic Targeting of TROP-2 in Treatment-Resistant Prostate Cancer. Clin. Cancer Res. 2023, 29, 2324–2335. [Google Scholar] [CrossRef]
  62. Guerra, E.; Relli, V.; Ceci, M.; Tripaldi, R.; Simeone, P.; Aloisi, A.L.; Pantalone, L.; La Sorda, R.; Lattanzio, R.; Sacchetti, A.; et al. Trop-2, Na+/K+ ATPase, CD9, PKCα, cofilin assemble a membrane signaling super-complex that drives colorectal cancer growth and invasion. Oncogene 2022, 41, 1795–1808. [Google Scholar] [CrossRef]
  63. Moretto, R.; Germani, M.M.; Giordano, M.; Conca, V.; Proietti, A.; Niccoli, C.; Pietrantonio, F.; Lonardi, S.; Tamburini, E.; Zaniboni, A.; et al. Trop-2 and Nectin-4 immunohistochemical expression in metastatic colorectal cancer: Searching for the right population for drugs’ development. Br. J. Cancer 2023, 128, 1391–1399. [Google Scholar] [CrossRef] [PubMed]
  64. Izci, H.; Punie, K.; Waumans, L.; Laenen, A.; Wildiers, H.; Verdoodt, F.; Desmedt, C.; Ardui, J.; Smeets, A.; Han, S.N.; et al. Correlation of TROP-2 expression with clinical-pathological characteristics and outcome in triple-negative breast cancer. Sci. Rep. 2022, 12, 22498. [Google Scholar] [CrossRef]
  65. Saber, H.; Leighton, J.K. An FDA oncology analysis of antibody-drug conjugates. Regul. Toxicol. Pharmacol. 2015, 71, 444–452. [Google Scholar] [CrossRef]
  66. 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]
  67. Bardia, A.; Messersmith, W.; Kio, E.; Berlin, J.; Vahdat, L.; Masters, G.; Moroose, R.; Santin, A.; Kalinsky, K.; Picozzi, V.; et al. Sacituzumab govitecan, a Trop-2-directed antibody-drug conjugate, for patients with epithelial cancer: Final safety and efficacy results from the phase I/II IMMU-132-01 basket trial. Ann. Oncol. 2021, 32, 746–756. [Google Scholar] [CrossRef]
  68. Hafeez, U.; Parakh, S.; Gan, H.K.; Scott, A.M. Antibody–drug conjugates for cancer therapy. Molecules 2020, 25, 4764. [Google Scholar] [CrossRef]
  69. Bardia, A.; Tolaney, S.M.; Punie, K.; Loirat, D.; Oliveira, M.; Kalinsky, K.; Zelnak, A.; Aftimos, P.; Dalenc, F.; Sardesai, S.; et al. Biomarker analyses in the phase III ASCENT study of sacituzumab govitecan versus chemotherapy in patients with metastatic triple-negative breast cancer. Ann. Oncol. 2021, 32, 1148–1156. [Google Scholar] [CrossRef]
  70. Loriot, Y.; Balar, A.V.; Petrylak, D.P.; Rezazadeh, A.; Grivas, P.; Flechon, A.; Jain, R.K.; Agarwal, N.; Bupathi, M.; Barthelemy, P.; et al. Efficacy of sacituzumab govitecan (SG) in locally advanced (LA) or metastatic urothelial cancer (mUC) by trophoblast cell surface antigen 2 (Trop-2) expression. J. Clin. Oncol. 2023, 41, 4579. [Google Scholar] [CrossRef]
  71. Rugo, H.; Bardia, A.; Marmé, F.; Cortés, J.; Schmid, P.; Loirat, D.; Trédan, O.; Ciruelos, E.; Dalenc, F.; Pardo, P.G.; et al. Abstract GS1-11: Sacituzumab Govitecan (SG) vs Treatment of Physician’s Choice (TPC): Efficacy by Trop-2 Expression in the TROPiCS-02 Study of Patients (Pts) With HR+/HER2– Metastatic Breast Cancer (mBC). Cancer Res. 2023, 83 (Suppl. S5), GS1-11. [Google Scholar] [CrossRef]
  72. Shimizu, T.; Sands, J.; Yoh, K.; Spira, A.; Garon, E.B.; Kitazono, S.; Johnson, M.L.; Meric-Bernstam, F.; Tolcher, A.W.; Yamamoto, N.; et al. First-in-Human, Phase I Dose-Escalation and Dose-Expansion Study of Trophoblast Cell-Surface Antigen 2-Directed Antibody-Drug Conjugate Datopotamab Deruxtecan in Non-Small-Cell Lung Cancer: TROPION-PanTumor01. J. Clin. Oncol. 2023, 41, 4678–4687. [Google Scholar] [CrossRef]
  73. Cardillo, T.M.; Rossi, D.L.; Zalath, M.B.; Liu, D.; Arrojo, R.; Sharkey, R.M.; Chang, C.-H.; Goldenberg, D.M. Predictive biomarkers for sacituzumab govitecan efficacy in Trop-2-expressing triple-negative breast cancer. Oncotarget 2020, 11, 3849–3862. [Google Scholar] [CrossRef]
  74. Trerotola, M.; Guerra, E.; Ali, Z.; Aloisi, A.L.; Ceci, M.; Simeone, P.; Acciarito, A.; Zanna, P.; Vacca, G.; D’Amore, A.; et al. Trop-2 cleavage by ADAM10 is an activator switch for cancer growth and metastasis. Neoplasia 2021, 23, 415–428. [Google Scholar] [CrossRef] [PubMed]
  75. Guerra, E.; Trerotola, M.; Relli, V.; Lattanzio, R.; Tripaldi, R.; Vacca, G.; Ceci, M.; Boujnah, K.; Garbo, V.; Moschella, A.; et al. Trop-2 induces ADAM10-mediated cleavage of E-cadherin and drives EMT-less metastasis in colon cancer. Neoplasia 2021, 23, 898–911. [Google Scholar] [CrossRef]
  76. Alberti, S.; Trerotola, M.; Guerra, E. The Hu2G10 mAb targets the cleaved-activated form of Trop-2 and exploits vulnerability of multiple human cancers. J. Clin. Oncol. 2022, 40 (Suppl. S16), e14548. [Google Scholar] [CrossRef]
  77. Alberti, S.; Trerotola, M.; Guerra, E. Abstract 340: The Hu2G10 tumor-selective anti-Trop-2 monoclonal antibody targets the cleaved-activated Trop-2 and shows therapeutic efficacy against multiple human cancers. Cancer Res. 2022, 82 (Suppl. S12), 340. [Google Scholar] [CrossRef]
  78. Kim, H.; Guerra, E.; Baek, E.; Jeong, Y.; You, H.; Yu, B.; Jang, T.; Saverio, A.; Chung, C.-W.; Park, C. Abstract 328: LCB84, a TROP2-targeted ADC, for treatment of solid tumors that express TROP-2 using the hu2G10 tumor-selective anti-TROP2 monoclonal antibody, a proprietary site-directed conjugation technology and plasma-stable tumor-selective linker chemistry. Cancer Res. 2022, 82 (Suppl. S12), 328. [Google Scholar]
  79. Guerra, E.; Trerotola, M.; Relli, V.; Lattanzio, R.; Tripaldi, R.; Ceci, M.; Boujnah, K.; Pantalone, L.; Sacchetti, A.; Havas, K.M.; et al. 3D-Informed Targeting of the Trop-2 Signal-Activation Site Drives Selective Cancer Vulnerability. Mol. Cancer Ther. 2023, 22, 790–804. [Google Scholar] [CrossRef]
  80. Guerra, E.; Trerotola, M.; Relli, V.; Lattanzio, R.; Ceci, M.; Boujnah, K.; Pantalone, L.; Di Pietro, R.; Iezzi, M.; Tinari, N.; et al. The 2EF Antibody Targets a Unique N-Terminal Epitope of Trop-2 and Enhances the In Vivo Activity of the Cancer-Selective 2G10 Antibody. Cancers 2023, 15, 3721. [Google Scholar] [CrossRef]
  81. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  82. Kumar, M.A.; Baba, S.K.; Sadida, H.Q.; Marzooqi, S.A.; Jerobin, J.; Altemani, F.H.; Algehainy, N.; Alanazi, M.A.; Abou-Samra, A.-B.; Kumar, R.; et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct. Target. Ther. 2024, 9, 27. [Google Scholar]
  83. Elsharkasy, O.M.; Nordin, J.Z.; Hagey, D.W.; de Jong, O.G.; Schiffelers, R.M.; Andaloussi, S.E.; Vader, P. Extracellular vesicles as drug delivery systems: Why and how? Adv. Drug Deliv. Rev. 2020, 159, 332–343. [Google Scholar] [CrossRef]
  84. Wei, H.; Chen, J.; Wang, S.; Fu, F.; Zhu, X.; Wu, C.; Liu, Z.; Zhong, G.; Lin, J. A Nanodrug Consisting Of Doxorubicin And Exosome Derived From Mesenchymal Stem Cells For Osteosarcoma Treatment In Vitro. Int. J. Nanomed. 2019, 14, 8603–8610. [Google Scholar] [CrossRef] [PubMed]
  85. Altanerova, U.; Jakubechova, J.; Benejova, K.; Priscakova, P.; Pesta, M.; Pitule, P.; Topolcan, O.; Kausitz, J.; Zduriencikova, M.; Repiska, V.; et al. Prodrug suicide gene therapy for cancer targeted intracellular by mesenchymal stem cell exosomes. Int. J. Cancer 2019, 144, 897–908. [Google Scholar] [CrossRef]
  86. Wu, A.; Han, M.; Ni, Z.; Li, H.; Chen, Y.; Yang, Z.; Feng, Y.; He, Z.; Zhen, H.; Wang, X. Multifunctional Sr/Se co-doped ZIF-8 nanozyme for chemo/chemodynamic synergistic tumor therapy via apoptosis and ferroptosis. Theranostics 2024, 14, 1939–1955. [Google Scholar] [CrossRef]
  87. Yong, T.; Zhang, X.; Bie, N.; Zhang, H.; Zhang, X.; Li, F.; Hakeem, A.; Hu, J.; Gan, L.; Santos, H.A.; et al. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat. Commun. 2019, 10, 3838. [Google Scholar] [CrossRef]
  88. Li, Y.; Gao, Y.; Gong, C.; Wang, Z.; Xia, Q.; Gu, F.; Hu, C.; Zhang, L.; Guo, H.; Gao, S. A33 antibody-functionalized exosomes for targeted delivery of doxorubicin against colorectal cancer. Nanomedicine 2018, 14, 1973–1985. [Google Scholar] [CrossRef] [PubMed]
  89. Saari, H.; Saari, H.; Lázaro-Ibáñez, E.; Viitala, T.; Vuorimaa-Laukkanen, E.; Siljander, P.; Yliperttula, M. Microvesicle- and exosome-mediated drug delivery enhances the cytotoxicity of Paclitaxel in autologous prostate cancer cells. J. Control. Release 2015, 220 Pt B, 727–737. [Google Scholar] [CrossRef]
  90. Niu, W.; Xiao, Q.; Wang, X.; Zhu, J.; Li, J.; Liang, X.; Peng, Y.; Wu, C.; Lu, R.; Pan, Y.; et al. A Biomimetic Drug Delivery System by Integrating Grapefruit Extracellular Vesicles and Doxorubicin-Loaded Heparin-Based Nanoparticles for Glioma Therapy. Nano Lett. 2021, 21, 1484–1492. [Google Scholar] [CrossRef] [PubMed]
  91. Zhang, Z.; Cao, W.; Xing, H.; Guo, S.; Huang, L.; Wang, L.; Sui, X.; Lu, K.; Luo, Y.; Wang, Y.; et al. A mix & act liposomes of phospholipase A2-phosphatidylserine for acute brain detoxification by blood–brain barrier selective-opening. Acta Pharm. Sin. B 2024, 14, 1827–1844. [Google Scholar]
  92. Feng, C.; Xiong, Z.; Wang, C.; Xiao, W.; Xiao, H.; Xie, K.; Chen, K.; Liang, H.; Zhang, X.; Yang, H. Folic acid-modified Exosome-PH20 enhances the efficiency of therapy via modulation of the tumor microenvironment and directly inhibits tumor cell metastasis. Bioact. Mater. 2021, 6, 963–974. [Google Scholar] [CrossRef]
  93. Hansen, H.P.; Trad, A.; Dams, M.; Zigrino, P.; Moss, M.; Tator, M.; Schön, G.; Grenzi, P.C.; Bachurski, D.; Aquino, B.; et al. CD30 on extracellular vesicles from malignant Hodgkin cells supports damaging of CD30 ligand-expressing bystander cells with Brentuximab-Vedotin, in vitro. Oncotarget 2016, 7, 30523–30535. [Google Scholar] [CrossRef]
  94. Barok, M.; Le Joncour, V.; Martins, A.; Isola, J.; Salmikangas, M.; Laakkonen, P.; Joensuu, H. ARX788, a novel anti-HER2 antibody-drug conjugate, shows anti-tumor effects in preclinical models of trastuzumab emtansine-resistant HER2-positive breast cancer and gastric cancer. Cancer Lett. 2020, 473, 156–163. [Google Scholar] [CrossRef]
  95. Andre, F.; Schartz, N.E.; Movassagh, M.; Flament, C.; Pautier, P.; Morice, P.; Pomel, C.; Lhomme, C.; Escudier, B.; Le Chevalier, T.; et al. Malignant effusions and immunogenic tumour-derived exosomes. Lancet 2002, 360, 295–305. [Google Scholar] [CrossRef]
  96. Trerotola, M.; Ganguly, K.K.; Fazli, L.; Fedele, C.; Lu, H.; Dutta, A.; Liu, Q.; De Angelis, T.; Riddell, L.W.; Riobo, N.A.; et al. Trop-2 is up-regulated in invasive prostate cancer and displaces FAK from focal contacts. Oncotarget 2015, 6, 14318–14328. [Google Scholar] [CrossRef] [PubMed]
  97. Barok, M.; Puhka, M.; Yazdi, N.; Joensuu, H. Extracellular vesicles as modifiers of antibody-drug conjugate efficacy. J. Extracell. Vesicles 2021, 10, e12070. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, X.; Zhang, H.; Bai, M.; Ning, T.; Ge, S.; Deng, T.; Liu, R.; Zhang, L.; Ying, G.; Ba, Y. Exosomes Serve as Nanoparticles to Deliver Anti-miR-214 to Reverse Chemoresistance to Cisplatin in Gastric Cancer. Mol. Ther. 2018, 26, 774–783. [Google Scholar] [CrossRef]
  99. Liang, G.; Zhu, Y.; Ali, D.J.; Tian, T.; Xu, H.; Si, K.; Sun, B.; Chen, B.; Xiao, Z. Engineered exosomes for targeted co-delivery of miR-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J. Nanobiotechnol. 2020, 18, 10. [Google Scholar] [CrossRef] [PubMed]
  100. Nandi, I.; Smith, H.W.; Sanguin-Gendreau, V.; Ji, L.; Pacis, A.; Papavasiliou, V.; Zuo, D.; Nam, S.; Attalla, S.S.; Kim, S.H.; et al. Coordinated activation of c-Src and FOXM1 drives tumor cell proliferation and breast cancer progression. J. Clin. Investig. 2023, 133, e162324. [Google Scholar] [CrossRef] [PubMed]
  101. Bankhead, A.; McMaster, T.; Wang, Y.; Boonstra, P.S.; Palmbos, P.L. TP63 isoform expression is linked with distinct clinical outcomes in cancer. EBioMedicine 2020, 51, 102561. [Google Scholar] [CrossRef]
  102. Zhang, Y.; Yan, W.T.; Yang, Z.Y.; Li, Y.L.; Tan, X.N.; Jiang, J.; Zhang, Y.; Qi, X.W. The role of WT1 in breast cancer: Clinical implications, biological effects and molecular mechanism. Int. J. Biol. Sci. 2020, 16, 1474–1480. [Google Scholar] [CrossRef]
  103. Zhao, M.; DiPeri, T.P.; Raso, M.G.; Zheng, X.; Rizvi, Y.Q.; Evans, K.W.; Yang, F.; Akcakanat, A.; Estecio, M.R.; Tripathy, D.; et al. Epigenetically upregulating TROP2 and SLFN11 enhances therapeutic efficacy of TROP2 antibody drug conjugate sacitizumab govitecan. NPJ Breast Cancer 2023, 9, 66. [Google Scholar] [CrossRef] [PubMed]
  104. Fu, C.; Yu, L.; Miao, Y.; Liu, X.; Yu, Z.; Wei, M. Peptide-drug conjugates (PDCs): A novel trend of research and development on targeted therapy, hype or hope? Acta Pharm. Sin. B 2023, 13, 498–516. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) The expression of TACSTD2 in various tumor tissues (red) and adjacent normal tissues (blue) was analyzed via the TIMER2.0 database (http://timer.cistrome.org/ (accessed on 10 May 2024)) (* p-value < 0.05; ** p-value < 0.01; *** p-value < 0.001). (B) Tumors with high Trop-2 expression (red: tumor tissue, grey: normal tissue; * p < 0.05) that could potentially benefit from Trop-2-targeted ADC therapeutics (GEPIA database http://gepia.cancer-pku.cn (accessed on 10 May 2024)).
Figure 1. (A) The expression of TACSTD2 in various tumor tissues (red) and adjacent normal tissues (blue) was analyzed via the TIMER2.0 database (http://timer.cistrome.org/ (accessed on 10 May 2024)) (* p-value < 0.05; ** p-value < 0.01; *** p-value < 0.001). (B) Tumors with high Trop-2 expression (red: tumor tissue, grey: normal tissue; * p < 0.05) that could potentially benefit from Trop-2-targeted ADC therapeutics (GEPIA database http://gepia.cancer-pku.cn (accessed on 10 May 2024)).
Pharmaceuticals 17 00652 g001
Figure 2. Mechanism of action of ADCs: (a) PH-sensitive linkers enable ADCs to release cytotoxic drugs in the acidic tumor microenvironment, where tumor cells generate a lot of lactic acid through glycolytic metabolism (known as the Warburg effect), allowing certain ADCs to exert anti-tumor effects without complete integration of the conjugates. (b) ADCs bind to extracellular vesicles (EVs) that are released by cancer cells expressing specific target proteins. These EVs serve as carriers, transporting the ADCs to nearby and distant cancer cells, resulting in anti-cancer effects and potentially altering the adverse effects associated with ADC treatment. (c) Liposoluble cytotoxic drugs can permeate the phospholipid bilayer, allowing them to be released from Ag+ cancer cells and impacting nearby Ag− cancer cells, ultimately playing an anti-tumor role.
Figure 2. Mechanism of action of ADCs: (a) PH-sensitive linkers enable ADCs to release cytotoxic drugs in the acidic tumor microenvironment, where tumor cells generate a lot of lactic acid through glycolytic metabolism (known as the Warburg effect), allowing certain ADCs to exert anti-tumor effects without complete integration of the conjugates. (b) ADCs bind to extracellular vesicles (EVs) that are released by cancer cells expressing specific target proteins. These EVs serve as carriers, transporting the ADCs to nearby and distant cancer cells, resulting in anti-cancer effects and potentially altering the adverse effects associated with ADC treatment. (c) Liposoluble cytotoxic drugs can permeate the phospholipid bilayer, allowing them to be released from Ag+ cancer cells and impacting nearby Ag− cancer cells, ultimately playing an anti-tumor role.
Pharmaceuticals 17 00652 g002
Table 1. Representative clinical trials related to Trop-2-targeted ADC combination therapy.
Table 1. Representative clinical trials related to Trop-2-targeted ADC combination therapy.
Combination TherapyIdentifierCombinePhaseRecruitment StatusCancer Type
Chemotherapy NCT06040970SG + cisplatinPhase 1/2RecruitingOvarian and endometrial cancer
NCT06065371SG + capecitabinePhase 1Not yet recruitingGastrointestinal cancers
NCT05089734SG + docetaxelPhase 3Active, not recruitingNSCLC *
NCT05555732Dato-DXd + pembrolizumab + platinumPhase 3RecruitingNSCLC
Molecularly targeted drugsNCT04039230SG + talazoparibPhase 1/2RecruitingBreast cancer
NCT05113966SG + talazoparibPhase 2Active, not recruitingTNBC *
NCT05143229SG + alpelisibPhase 1RecruitingBreast cancer
NCT04826341SG + berzosertibPhase 1RecruitingSolid tumor
NCT03992131SG + rucaparibPhase 1/2TerminatedSolid tumor
NCT05006794SG + GS-9716Phase 1RecruitingSolid tumor
NCT05675579Dato-DXd + osimertinibPhase 3Not yet recruitingNSCLC
NCT03944772Dato-DXd + osimertinibPhase 2Not yet recruitingNSCLC
NCT05417594Dato-DXd + AZD9574Phase 1/2RecruitingSolid tumor
ImmunotherapyNCT05675579SG + pembrolizumabPhase 2RecruitingTNBC
NCT06055465SG + pembrolizumabPhase 2RecruitingNSCLC
NCT05633654SG + pembrolizumabPhase 3RecruitingTNBC
NCT04468061SG + pembrolizumabPhase 2RecruitingTNBC
NCT05609968SG + pembrolizumabPhase 3RecruitingNSCLC
NCT05382286SG + pembrolizumabPhase 3RecruitingTNBC
NCT06081244SG + pembrolizumabPhase 2RecruitingTNBC
NCT03869190SG + atezolizumabPhase 1/2RecruitingBladder cancer
NCT04434040SG + atezolizumabPhase 2Active, not recruitingTNBC
NCT03424005SG + atezolizumabPhase 1/2RecruitingBreast cancer
NCT06133517SG + zimberelimab (ZIM) + domvanalimab (DOM)Phase 2Not yet recruitingMIBC *
NCT05535218SG + pembrolizumabPhase 2Enrolling by invitationMIBC
NCT06161532SG + atezolizumabPhase 2Not yet recruitingRare genitourinary tumors
NCT04863885SG + ipilimumab + nivolumabPhase 1/2Active, not recruitingUrothelial cancer
NCT03337698SG + atezolizumabPhase 1/2RecruitingNSCLC
NCT03971409SG + avelumabPhase 2RecruitingTNBC
NCT04143711SG + DF1001Phase 1/2RecruitingSolid tumor
NCT05633667SG + zimberelimab (ZIM) + domvanalimab (DOM)Phase 2RecruitingNSCLC
NCT06103864Dato-DXd + durvalumabPhase 3RecruitingTNBC
NCT05215340Dato-DXd + pembrolizumabPhase 3RecruitingNSCLC
NCT05629585Dato-DXd + durvalumabPhase 3RecruitingTNBC
NCT06112379Dato-DXd + durvalumabPhase 3RecruitingBreast cancer
NCT06357533Dato-DXd + rilvegostomigPhase 3Not yet recruitingNSCLC
NCT03742102Dato-DXd + durvalumabPhase 1/2RecruitingTNBC
NCT06328387SG + hydroxychloroquinePhase 1/2RecruitingBreast cancer
ChemoimmunotherapyNCT05555732Dato-DXd + pembrolizumab + platinumPhase 3Recruiting1L NSCLC
NCT05489211Dato-DXd + saruparib (AZD5305) + carboplatinPhase 2RecruitingSolid tumors
NCT04526691Dato-DXd + pembrolizumab + platinumPhase 1RecruitingAdvanced or metastatic NSCLC
NCT04612751DatoDXd + durvalumab + carboplatinPhase 1RecruitingAdvanced or metastatic NSCLC
NCT05687266DatoDXd + durvalumab + carboplatinPhase 3RecruitingAdvanced
NSCLC without AGAs *
NCT05061550Dato-DXd + durvalumab + platinumPhase 2RecruitingNSCLC
OthersNCT06328387SG + hydroxychloroquinePhase 1/2RecruitingBreast cancer
NCT06100874SG + trastuzumabPhase 2RecruitingBreast cancer
NCT06244485SG + valemetostat tosylatePhase 1RecruitingSolid tumor
NCT04724018 SG + EVPhase 1Active, not recruitingUrothelial cancer
NCT03725761SG + enzalutamide/abiraterone/ARN-509Phase 2RecruitingProstate cancer
NCT06238921SG + zimberelimab with SRS *Phase 1/2Not yet recruitingmTNBC * with brain metastases
NCT05833867SG + adaptive radiation therapyPhase 1RecruitingMIBC
NCT05520723SG + loperamide + G-CSFPhase 2Active, not recruitingTNBC
NCT06167317SG + GS-0201Phase 1RecruitingSolid tumor
* NSCLC, non-small cell lung cancer; (m)TNBC, (metastatic) triple-negative breast cancer; MIBC, muscle-invasive bladder cancer; AGAs, actionable genomic alterations; SRS, stereotactic radiation. Data were collected up to 20 April 2024 from ClinicalTrials.gov.
Table 2. Representative clinical trials related to biomarkers of Trop-2-Targeted ADCs.
Table 2. Representative clinical trials related to biomarkers of Trop-2-Targeted ADCs.
IdentifierStudy TypeConditionsStatus
NCT06240195ObservationalTNBCRecruiting
NCT06341478InterventionalMIBCRecruiting
NCT05097599InterventionalSolid tumorRecruiting
NCT05770531InterventionalBreast cancerSuspended
NCT05332561InterventionalBreast cancerRecruiting
NCT06311214InterventionalSolid tumorNot yet recruiting
NCT06178588InterventionalCholangiocarcinomaRecruiting
Data were collected up to 20 April 2024 from ClinicalTrials.gov.
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

Jiang, Y.; Zhou, H.; Liu, J.; Ha, W.; Xia, X.; Li, J.; Chao, T.; Xiong, H. Progress and Innovative Combination Therapies in Trop-2-Targeted ADCs. Pharmaceuticals 2024, 17, 652. https://doi.org/10.3390/ph17050652

AMA Style

Jiang Y, Zhou H, Liu J, Ha W, Xia X, Li J, Chao T, Xiong H. Progress and Innovative Combination Therapies in Trop-2-Targeted ADCs. Pharmaceuticals. 2024; 17(5):652. https://doi.org/10.3390/ph17050652

Chicago/Turabian Style

Jiang, Yizhi, Haiting Zhou, Junxia Liu, Wentao Ha, Xiaohui Xia, Jiahao Li, Tengfei Chao, and Huihua Xiong. 2024. "Progress and Innovative Combination Therapies in Trop-2-Targeted ADCs" Pharmaceuticals 17, no. 5: 652. https://doi.org/10.3390/ph17050652

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