International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Ovarian cancer is the second most lethal gynecologic malignancy, with an estimated 206,839 mortalities associated with the disease in 2022 according to the recent global cancer statistics (1). The data from the American Cancer Society in 2024 indicated that there has been no improvement in the mortality/incidence rate of 62% of ovarian cancers in the last 30 years (2). More than 80% of patients with ovarian cancer experience recurrence and eventually develop treatment resistance after surgery and platinum-based chemotherapy (3,4). The mainstay of treatment for platinum-resistant ovarian cancer (PROC), which progresses within 6 months of completing the platinum treatment, has been the sequential use of cytotoxic drugs, including pegylated liposomal doxorubicin (PLD), gemcitabine, paclitaxel and topotecan, in recent years (4). However, the effective proportion of these drugs is only 10–15%, and the expected survival time is ~12 months (5,6). The ongoing research on developing new drugs and identifying new therapeutic targets is constantly evolving.
ADCs selectively deliver cytotoxic payloads to tumor cells, representing a rapidly evolving cancer therapy (6–8). At present, 15 ADCs have been approved by the US Food and Drug Administration for tumor treatment, and the development of these drugs for PROC has gained interest. In November 2022, mirvetuximab soravtansine (MIRV; ELAHERE®) was approved by the US Food and Drug Administration (FDA) as the first ADC for ovarian cancer (9). MIRV targets folate receptor α (FRα) on the surface of tumor cells to induce tumor cell killing, thereby displaying promising clinical activity in patients with FRα-positive ovarian cancer (10). Other ADCs are still under active development in the treatment of PROC. In addition, studies on using ADCs in combination therapy with various anticancer drugs are underway (NCT05887609, NCT06660511, NCT05941507).
The present review aimed to describe the structure and mechanism of action of ADCs, present an overview of the progress in the effectiveness of various types of ADCs in treating PROC in preclinical studies and clinical trials, and to discuss the recent advances in combining ADCs with chemotherapeutic agents, targeted agents and immunotherapies for PROC treatment. The present study also analyzed the toxicity characteristics and management of ADCs and finally considered the future development of ADCs for ovarian cancer.
A literature search was performed using the Web of Science (https://www.webofscience.com/wos/) and PubMed (https://pubmed.ncbi.nlm.nih.gov/) databases. ‘ADC’, ‘monotherapy’, ‘combination therapy’, ‘platinum-resistant ovarian cancer’ and ‘research progress’ were used as the core search terms. Each core search term was searched separately, and then Boolean logical operators ‘AND’ and ‘OR’ were applied for combined searches, for example, ‘(ADC AND monotherapy AND platinum-resistant ovarian cancer) OR (ADC AND combination therapy AND platinum-resistant ovarian cancer)’. The search fields were limited to the title, abstract and keywords to pinpoint relevant literature. The inclusion criteria were as follows: i) Studies focused on ADC monotherapy or combination therapy for PROC; ii) clinical research, basic research or review articles; iii) language limited to English; and iv) published within the last 10 years (January 1, 2014 to December 1, 2024) to obtain the latest research developments. The literature needed for the writing process was also included where appropriate. The exclusion criteria were as follows: i) Duplicate publications or highly similar content; ii) non-research literature such as conference abstracts, reviews, letters, news reports and so forth; and iv) literature with weak relevance or missing key data.
The quality of clinical studies was assessed by evaluating randomization methods, implementation of blinding, and descriptions of loss of visits and withdrawals; the assessment for the quality of basic research covered dimensions such as rationality of experimental design, sample size and statistical methods of data; and that of review articles focused on the comprehensiveness of their literature coverage, depth of analysis and logic. Two professionals performed independent assessment, and a third expert was asked to adjudicate in case of disagreement.
ADCs are complexes comprising an antibody, a cytotoxic payload and a connecting linker. They specifically recognize tumor cell surface antigens, induce endocytosis and release cytotoxic drugs into tumor cells, ultimately leading to the death of tumor cells (6,11). The connection of cytotoxic drugs with antibodies was first realized in the 1950s (12). Researchers conducted clinical trials on the mouse immunoglobulin G (IgG)-based ADCs in the 1980s (13). Following a long and slow development, the field has become active again in the last decade, with ADCs entering the global market at a much faster pace. Usually, ADCs are administered intravenously. Then, the antibodies of ADCs bind to the antigen on cancer cells, penetrate the cells via receptor-mediated endocytosis to form endosomes and fuse with lysosomes. The cytotoxic payload detaches from the antibody and diffuses into the cell in the presence of various lysosome enzymes, resulting in tumor cell death by disrupting DNA structures or inhibiting microtubule polymerization (8,14,15). In addition, the payload can penetrate the extracellular matrix and surrounding cells, resulting in a bystander effect (16). Fig. 1 shows a schematic representation of the structure and mechanism of action of conventional ADCs.
The most commonly used antibody backbone in ADCs is IgG. The antibody-binding antigen should be highly expressed on the surface of tumor cell membranes with minimal expression on normal tissues (17,18) to enhance the effectiveness of the drug by delivering it to specific targets, protect the non-target tissues from the damage caused by chemotherapy, and reduce the systemic toxicity. FRα, human epidermal growth factor receptor 2 (HER2), trophoblast cell surface antigen-2 (TROP2), mesothelin, sodium-dependent phosphate transporter 2b (NaPi2b) and CDH6 are usually overexpressed in epithelial ovarian cancer, making them the most commonly used conjugated antigens (19).
The covalent binding of antigen-targeting antibodies to cytotoxic payloads requires connecting linkers (20). Linkers have two main functions; the primary one is maintaining stability in the bloodstream, keeping the cytotoxic payload attached to the antibody when it moves through the plasma; the other aspect is to efficiently release the payload into the tumor (8). The linkers should keep this attachment stable and unchanged in the bloodstream, with the drug being released only after antigen-antibody binding (21). Linkers are cleavable or non-cleavable depending on their chemical characteristics. Cleavable linkers are chemically unstable structures that can deliver drugs extracellularly and induce the killing of nearby tumor cells (22,23). The non-cleavable linker releases the drug solely when the antibody undergoes internalization and degradation within the lysosomes of the target cells. However, the cytotoxic payloads are released after the apoptosis of tumor cells, which may also result in the non-specific killing of surrounding tumor cells (24).
The cytotoxic payloads in ADCs now encompass a range of DNA-targeting agents, microtubule-binding proteins and some topoisomerase 1 inhibitors. The agents targeting microtubules are the most frequently used payloads, targeting the medenosine and periwinkle alkaloid sites. They interfere with the kinetics of microtubule protein polymerization and maintain the cell cycle in the G2/M phase, leading to cell death (25,26). Not all tumor types are sensitive to a given type of payload and, therefore, diversifying payloads is critical to expanding the indications of ADCs.
Alternative strategies based on the use of ADCs are highly effective in treating cancer. The use of ADCs in clinical oncology has increased with the advent of novel technologies and the discovery of new targets (9,27). To date, 15 ADCs have received marketing approval from the FDA, and hundreds more are undergoing preclinical and clinical evaluation (28). Besides hematological tumors, ADCs have been approved for use in breast, gastric, lung, uroepithelial, cervical, ovarian and head-and-neck squamous cell carcinomas. Several previous studies have described the ADC drugs approved for oncology treatment (29–31).
Solid tumors offer a broad opportunity for ADC development. The treatment choices for advanced or metastatic recurrent solid tumors are limited, and current immunotherapies rarely cure the disease (9). Therefore, ADCs have immense potential and unique advantages in a wide range of solid tumors.
FRα, one of the four members of the FR family, is a glycosylphosphatidylinositol-anchored membrane protein (32) that binds to folic acid and related compounds and transports them into the cell through receptor-mediated endocytosis (33,34). FRα is seldom expressed in normal tissues but is specifically expressed in epithelial tumors, including ovarian cancer, endometrial cancer, triple-negative breast cancer and non-small-cell lung cancer (35,36), making FRα a promising target for tumor therapy.
In November 2022, MIRV was approved by the FDA as the first ADC for ovarian cancer (37). Targeting FRα, MIRV comprises an antibody, a cleavable linker and a small-molecule microtubule inhibitor DM4 (a maytansine derivative) (38). MIRV is endocytosed by tumor cells and transferred to lysosomes, where it is degraded to release lysine-DM4. Lysine-DM4 produces metabolites that can inhibit tubulin polymerization and microtubule assembly, causing cell cycle arrest and leading to cell death (39). Further, the disintegration of tumor cells leads to the release of catabolic metabolites, which may induce bystander killing (40–42).
Several recent clinical studies have been conducted on the efficacy and safety of MIRV in ovarian cancer (43–45). A phase II trial (NCT04296890) including 106 patients with PROC showed a reduction in tumor size in 71.4% of patients, an objective response rate (ORR) of 32.4% and a median duration of remission (DOR) of 6.9 months (43). This result supports the clinically meaningful efficacy of MIRV in PROC expressing FRα, regardless of prior therapy or sequencing. Recently, a phase III trial (NCT04209855) compared MIRV with chemotherapeutic drugs (paclitaxel, PLD or topotecan) (45). The results showed a median progression-free survival (PFS) of 5.62 months in the MIRV group and 3.98 months in the chemotherapy group. The ORR of MIRV (42.3%) was significantly higher compared with that of chemotherapy (15.9%). The median overall survival of MIRV (16.46 months) was also significantly longer compared with that of chemotherapy (12.75 months). Fewer adverse events also demonstrated the safety of MIRV over chemotherapy. These results support the use of MIRV for treating platinum-resistant epithelial ovarian cancer, providing a new alternative for treating PROC (46). Some ongoing clinical trials are assessing the efficacy and adverse effects of MIRV in PROC (NCT06365853, NCT06682988 and NCT05622890).
Besides MIRV, luveltamab tazevibulin (STRO-002) is the second ADC drug targeting FRα. STRO-002 acts as a tumor killer by releasing the tubulin-targeting cytotoxin 3-aminophenyl hemiasterlin (SC209) and reducing the potential for drug efflux (47). STRO-002 is still in phase I–III trials (NCT03748186, NCT05200364, NCT06238687 and NCT05870748), both as a monotherapy or in combination with bevacizumab in a variety of solid tumors, including PROC.
Farletuzumab ecteribulin (MORAb-202) is another ADC that targets FRα-expressing tumor cells. MORAb-202 initially inhibited tumor growth in a patient-derived xenograft (PDX) model of triple-negative breast cancer, demonstrating durable efficacy proportional to FRα expression (48). In clinical trials, MORAb-202 showed promising antitumor activity and good tolerability in a phase I study (NCT03386942) of FRα-positive solid tumors, including PROC (49). Also, two phase I/II trials of MORAb-202 for treating PROC (NCT05613088 and NCT04300556) are underway.
HER2 is another promising target that can enhance cell proliferation, differentiation and migration and inhibit apoptosis (50). HER2 is upregulated in a range of solid tumors, including breast, gastric and gynecological tumors (51). However, HER2-targeted therapies are not yet approved for use in diseases other than breast, lung, gastric and colorectal cancers (52). Trastuzumab deruxtecan (T-DXd) targets HER2 and contains a topoisomerase I inhibitor payload (53). T-DXd is approved for treating HER2-expressing breast and gastric cancers and HER2-mutated non-small-cell lung cancer (50). Preclinical studies demonstrated the antitumor activity of T-DXd against primary and metastatic ovarian tumors overexpressing HER2 (54). A global multicenter phase II study (NCT04482309) was conducted to evaluate the efficacy and safety of T-DXd in patients with HER2-expressing solid tumors across seven cohorts, including three major gynecological tumors. T-DXd treatment exhibited robust clinical efficacy, conferring sustained clinical benefits for patients with HER2-expressing solid tumors (55). Among all studied tumor types, the highest ORR was observed in the three major gynecological tumor cohorts, with an ORR of 57.5% for endometrial cancer, 50.0% for cervical cancer and 45.0% for ovarian cancer. Among the ovarian cancer cohort, 35.0% of the patients had previously received five or more lines of treatment and the median overall survival time was 13.2 months; by contrast, the median overall survival time for patients with high HER2 expression (IHC 3+) increased to 20.0 months (55). These findings indicate that T-DXd is a promising drug for HER2-expressing recurrent PROC (56).
TROP2 is another tumor target highly expressed in ovarian cancer. This is a type I cell surface glycoprotein first discovered in human trophoblasts (57). It is upregulated in a variety of malignant tumors and plays an important role in tumor development, invasion and metastasis (58). Datopotamab deruxtecan (Dato-DXd; DS-1062a), an effective topoisomerase I inhibitor, is an ADC that targets TROP2. Dato-DXd specifically binds to TROP2, transports it intracellularly to lysosomes and releases DXd. In preclinical experiments, Dato-DXd induced DNA damage and apoptosis of tumor cells in vitro and displayed antitumor activity in vivo in xenograft tumors with high TROP2 expression (59). When mixed with TROP2 (IHC 3+) tumor cells, Dato-DXd showed a significant bystander-killing effect on tumor cells with low TROP2 expression, thus prolonging the survival time of epithelial ovarian cancer xenograft models and reducing toxicity (60). Most clinical trials are conducted on breast cancer. A phase II clinical trial (NCT05489211) is currently evaluating Dato-DXd for treating advanced/metastatic solid tumors, including ovarian cancer (61). The efficacy of Dato-DXd in ovarian cancer expressing TROP2 deserves continued attention.
Mesothelin represents a promising potential target for ovarian cancer. High expression of mesothelin is associated with chemotherapeutic resistance and poor prognosis in epithelial ovarian cancer (62). Anetumab ravtansine exhibits a high degree of affinity for mesothelin, using the microtubule inhibitor DM4 as a payload. It has displayed high antitumor activity and good tolerability as a monotherapy in the preclinical models of ovarian cancer (63). A phase I study (NCT02751918) determined the antitumor activity, safety and pharmacokinetics of anetumab ravtansine in PROC-expressing mesothelin. The result showed an ORR of 27.7%, DOR of 7.6 months and PFS of 5.0 months (64). This result suggests that targeting mesothelin is an effective and well-tolerated treatment option in patients with PROC. The development of other combination therapy regimens for anetumab ravtansine (65) and other ADCs targeting mesothelin (BMS-986148) (66) has also provided preliminary evidence of clinical activity and tolerable adverse reactions and toxicity in ovarian cancer.
NaPi2b is another promising target for ADC, which is expressed in 95% of ovarian cancers (67). Lifastuzumab vedotin (LIFA) targeting NaPi2b displayed activity and acceptable safety in phase I studies (68,69). A phase II study compared the efficacy of LIFA and polyethylene glycol liposome doxorubicin in patients with PROC. The results showed that LIFA was well-tolerated, and ORR increased (34 vs. 15%; P=0.03) (70). Moreover, XMT-1536 and TUB-040 are other ADCs targeting NaPi2b. Currently, the safety and efficacy of these drugs in PROC are under evaluation in clinical trials (NCT06517485, NCT03319628, NCT06517433 and NCT06303505).
New ADCs for ovarian cancer are constantly being developed. Raludotatug deruxtecan (R-DXd) targets cadherin 6 (CDH6) and was effective and safe in a preclinical study using PDX tumor models of serous ovarian cancer expressing CDH6 (71). B7-H4-directed ADC, using a pyrrolobenzodiazepine-dimer payload, displayed antitumor activity in a PARP inhibitor and platinum-resistant PDX model of high-grade serous ovarian cancer (72).
The findings of several newly completed clinical trials examining the use of ADCs in ovarian cancer (NCT06517485 and NCT06517433) are yet to be published. Table I lists the clinical trials investigating ADC monotherapy for ovarian cancer that deserve continued attention in the future.
Chemotherapeutic agents such as platinum, which can damage DNA, target the S phase of the cell cycle and induce the G2/M phase. Preclinical studies described below have shown that these chemotherapeutic agents can bind effectively to ADCs containing microtubule-disrupting payloads. MIRV has been demonstrated to arrest the cell cycle and enhance DNA damage. The combination of MIRV with carboplatin, Adriamycin or doxorubicin has been found to inhibit proliferation synergistically in ovarian cancer cell lines and PDX models (73). The combination therapy of carboplatin and STRO-002 further improved the efficacy of STRO-002 (47). The combination of anetumab ravtansine with carboplatin or PLD demonstrated enhanced efficacy in vivo and in vitro (in ovarian cancer cell line and PDX models) compared with monotherapy in ovarian cancer (74). MIRV combined with carboplatin in early clinical trials achieved an ORR of 71%, with only minor adverse events (75), making it a highly active therapy for ovarian cancer.
A prevalent issue in solid tumors is inadequate blood flow and hypoxia within the vasculature (76). Using drugs such as anti-vascular endothelial growth factor antibodies (e.g., bevacizumab) can modulate angiogenesis and vascular porosity, thereby altering the tumor vascular system. Bevacizumab, the first targeted drug indicated for ovarian cancer, has been approved for use in combination with chemotherapy (77). Administering bevacizumab in combination with platinum-based chemotherapy after disease progression still improved PFS (78). ADCs have also shown safety and efficacy in combination with bevacizumab, particularly in PROC. Preclinical studies demonstrated that MIRV combined with bevacizumab resulted in rapid destruction of the tumor microvascular system and extensive areas of necrosis (73,74). Moreover, it was highly effective in a PDX model of PROC, causing significant regression and complete remission (73). Anetumab ravtansine combined with bevacizumab also demonstrated enhanced antitumor efficacy (74). These results emphasized the superior biological activity of the combination and further advanced the combination into clinical trials.
In clinical trials, the combination of MIRV and bevacizumab demonstrated efficacy and was well-tolerated in patients with PROC (79). In another cohort from the same study (NCT02606305), 94 patients with PROC treated with MIRV and bevacizumab had an ORR of 44%, an median duration of remission (mDOR) of 9.7 months, and an median progression-free survival (mPFS) of 8.2 months (80). Furthermore, incorporating MIRV into dual therapy with platinum and bevacizumab demonstrated enhanced efficacy and safety profiles in platinum-sensitive ovarian cancer (81). All these results point to the importance of ADCs combined with bevacizumab. Currently, a number of ongoing trials are investigating several additional combinations of different ADCs with bevacizumab (NCT05445778, NCT05200364 and NCT03587311).
Other than directly inducing cancer cell death using cytotoxic payloads, ADC also has antitumor immune activity (8,82,83). The related mechanisms, including Fc-mediated effector function, immune cell death, dendritic cell maturation, enhancement of T-cell infiltration and enhancement of immune memory, among others, have been the subject of discussion (84–87). ADCs have been shown to possess considerable immunomodulatory capacity in animal models (88). Combination of ADCs with immunotherapy in refractory tumors is a promising strategy for clinical treatment; its improved antitumor effects have been initially demonstrated in many preclinical studies and early clinical trials.
Immune checkpoint inhibitors (ICIs) combined with ADCs have synergistic effects in triple-negative breast cancer (89); the rationale behind this combination is that ADCs activate the immune system, whereas ICIs remove the brakes, allowing the immune system to regain its ability to recognize and kill tumor cells (90). Durvalumab combined with T-DXd (NCT03742102) exhibited an ORR of 100% and was found to be safe in a small group of patients with low expression of HER2. The interim results of T-DXd alone and combined with patulizumab (NCT04538742) showed ORRs of 77.3 and 82.0% and 12-month PFSs of 77.3 and 89.4%, respectively, demonstrating promising efficacy and controllable safety. The clinical benefits of ADCs combined with immunotherapy have been observed in clinical trials on both uroepithelial cancer (NCT03288545 and NCT04264936) and lung cancer (NCT02099058), besides breast cancer (91). MIRV combined with other agents, including pembrolizumab, was evaluated in FRα-positive PROC. Preliminary data showed good tolerability and therapeutic activity, with an ORR of 43%, mDOR of 6.9 months, and mPFS of 5.2 months for MIRV combined with pembrolizumab (NCT02606305). The synergistic effect of ADCs and immunotherapy can overcome treatment resistance, displaying encouraging efficacy and safety in tumor treatment. However, large, randomized phase III clinical trials are needed to test their efficacy against conventional therapy.
Table II lists the ongoing clinical trials on ADC combination therapy for ovarian cancer.
ADC has fewer side effects compared with conventional chemotherapy due to its ability to target tumor cells. The toxicity of ADCs arises primarily from the payload and secondarily from linker and bystander effects (92).
A common treatment-related adverse event (TRAE) associated with MIRV is ocular toxicity. A systematic review and meta-analysis showed that the most common TRAEs in patients treated with MIRV were blurred vision (all grades, 45%; grade III, 2%; no grade IV), nausea (all grades, 42%; grade III, 1%; no grade IV) and diarrhea (all grades, 42%; grade III, 2%; no grade IV) (93). In a previously mentioned phase II trial of MIRV (NCT04296890), the most common TRAEs were blurred vision (all grades, 41%; grade III, 6%; no grade IV), keratoconus (all grades, 29%; grade III, 8%; grade IV, 1%) and nausea (all grades, 29%; no grade III or grade IV) (43). Further, ocular events requiring dose reductions occurred in 12 patients (11%). One patient required discontinuation of therapy.
T-DXd is associated with interstitial lung disease (ILD)/pneumonia due to the expression of HER2 in lung epithelial cells (94). It is mainly localized to alveolar macrophages; the incidence and severity of ILD/pneumonia depend on the T-DXd dose and frequency of administration, suggesting that this ILD/pneumonia may be caused by cytotoxic lung injury (95). It is mild in most cases and can be effectively treated, but may be fatal in some cases (96). Respiratory disease (pneumonia) has been the most common cause of treatment-related mortalities in all types of ADC therapy (97). Other TRAEs include nausea and diarrhea, which are effectively controlled using antiemetic and antidiarrheal medications.
Toxicity management includes implementing supportive measures, suspension of treatment, dosage adjustments, or permanent discontinuation of therapy. Most ocular adverse events are mild and reversible; they improve or subside upon discontinuation or therapy improvement (98). Scheduling regular eye examinations to monitor early signs and symptoms, detecting ocular adverse effects in an early stage and prompting pharmacological intervention, promoting the prophylactic use of corticosteroids and lubricating eye drops, adjusting ADC dosage when needed, and maintaining clear communication with the ophthalmologist can help relieve symptoms before the vision is affected, ultimately helping the patient to continue treatment (98,99).
New guidelines for T-DXd-associated ILD/pneumonia toxicity have been published. A multidisciplinary team and timely management with steroids are recommended (100). Careful monitoring by a multidisciplinary team facilitates early detection [e.g., grade I (100)] of ILD/pneumonia, leading to timely discontinuation of medication and initiation of steroids, which prevents the development of fatal ILD/pneumonia.
The development of ADCs has some challenges. The target expression is not uniform in tumors due to the heterogeneity of tumor cells, and ADCs have limited effects on tumor cells with low or no target expression. Normal tissues expressing the target are attacked by the drug, limiting the drug dose. When combining drugs, the optimal dose of each drug and the order of administration also need to be re-explored. Other challenges include tumor cells developing resistance to ADCs through multiple mechanisms, the need to identify potential biomarkers for patient selection and limitations in current clinical trial designs.
ADC resistance can be caused by various mechanisms, including change in antigen expression, failure of ADC transportation, failure of ADC internalization, change in tumor sensitivity, exocytosis of ADC payload and activation of signaling pathways (101–103). Some of these mechanisms are not fully understood.
First, alterations occur in target antigen expression, including target downregulation, loss or mutations in target genes. Some tumor cells may evade ADC recognition and binding by reducing or completely losing target expression. Mutations in the target gene may also alter the structure and function of the target, thus affecting the binding affinity of the ADC to the target (101,102).
Second, the abnormal expression or defective function of endocytosis-related proteins, such as cell surface grid proteins, in some tumor cells can lead to the inability of ADC to enter the cell effectively during the internalization and processing of ADC. ADCs that enter the cell are usually transported to the lysosome for degradation, releasing cytotoxic drugs to play a role. If tumor cells enhance the stability of lysosomes or change the microenvironment of lysosomes, ADCs cannot release drugs in lysosomes, resulting in drug resistance (103,104).
Furthermore, a resistance mechanism related to drug metabolism and clearance involves high expression of drug efflux pumps (e.g., drug transporter proteins such as multidrug resistance protein 1 and multidrug resistance-associated protein 1), which can pump ADCs or their released cytotoxic drugs out of the cell, decreasing the drug concentration in the cell, thus leading to drug resistance (104). In breast cancer, acquired resistance to ADC developed in two different cancer cells after months of drug treatment, primarily due to increased expression of the drug efflux protein ABCC1 or decreased expression of HER2 antigen (105).
Finally, the alterations in multiple signaling pathways in tumor cells are also closely related to ADC resistance. For example, the activation of the PI3K/Akt/mTOR signaling pathway can promote the proliferation, survival and metabolism of tumor cells, making them resistant to the cytotoxic effects of ADCs. The abnormal activation of the JAK/STAT signaling pathway may also lead to the development of ADC resistance in tumor cells (106). Alterations in some signaling pathways involved in regulating the cell cycle and apoptosis (such as aberrant activation of STAT3) can also lead to the development of ADC resistance (106).
The selection of potential biomarkers for ADC drug development is also challenging. Normal tissues and cells may also partially express ADC targets, and therefore ADC payloads may have serious side effects on these normal cells (15). The biomarker should preferably be present only on tumor cells or tumor tissue and not expressed in normal cells and tissues. Uniform criteria to determine the expression level that accurately predicts ADC efficacy are lacking. For example, in the case of gosatuzumab, the TROP-2 ADC drug, although TROP-2 expression is associated with efficacy, the exact threshold of expression needs to be further defined with more sufficient evidence. In addition, the biomarker expression of different cells within the same tumor may vary or the biomarker expression levels of the same types of tumors originating from different patients may be different due to the existence of intra-tumor and inter-tumor heterogeneity. All of these increase the difficulty in finding universal biomarkers (107), making the selection of specific targets highly challenging. Moreover, the tumor cells lacking a specific target can proliferate rapidly in the presence of ADCs, leading to a new round of recurrence and drug resistance (108), again demonstrating the importance of finding tumor-specific targets.
Current clinical trials are mostly limited to the efficacy of ADC monotherapy. The information on the safety and toxicity of monotherapy and the efficacy of combination therapy is still in the preliminary stages. First, like-for-like comparisons of newer ADC drugs with standard-of-care or other comparable ADC drugs in clinical trials are scarce, limiting their status and superiority in their class and creating difficulties in dosing choices for clinicians and patients. Second, the maximum tolerated dose is usually derived from safety data in phase I trials. However, the cumulative toxicity of a drug may not be apparent until after multiple courses of therapy, which needs further investigation. In addition, the complexity of the design of sequencing, dosage and duration of combination therapy, as well as the lack of established theoretical and practical guidance, makes the design and implementation of clinical trials more difficult.
A number of other novel approaches to tumor treatment are currently available, which include tumor cell therapy, chimeric antigen receptor T-cell therapy (109), T-cell receptor engineered T-cell therapy (110), tumor-infiltrating lymphocyte therapy (111), ICI therapies, therapies using gene editing techniques such as CRISPR-Cas9 (112) and oncolytic virotherapy (113). ADCs have shown good efficacy in treating various hematological malignancies and solid tumors, with relatively good safety, controllable adverse effects and fewer serious adverse effects such as cytokine release syndrome (114). The precise delivery mechanism, excellent therapeutic effect and safety of ADC indicate its great developmental potential in the field of tumor therapy and wide recognition in the tumor therapy market.
However, the development of ADCs faces several practical challenges in clinical practice. First, comprehensive patient stratification from both clinical and molecular characteristics is needed for specific clinical applications. Besides determining the molecular typing of the patient's tumor cells to select sensitive ADC drugs, the patient's general conditions, such as age, physical status and comorbidities, and other tumor-related characteristics, such as tumor site, size, stage and metastasis, need to be comprehensively considered. For example, the tolerability of ADC therapy may need to be more carefully evaluated in patients with advanced stage and poor physical status. Further, patient tumor samples can be comprehensively analyzed using multi-omics technologies, such as genomics, transcriptomics, proteomics and metabolomics, to mine highly specific and sensitive biomarker combinations for individualized and precise treatment. Second, as ADCs are expensive, a study suggested a reduction in total drug acquisition costs by at least 50% to make it a cost-effective strategy (115). The present study recommends using advanced computer simulation and artificial intelligence techniques to improve drug design and screening efficiency, optimize research and development processes, and enhance production by increasing coupling efficiency and product purity, ultimately reducing research and development and manufacturing costs.
Treatment of PROC remains a major challenge. Despite the development of systemic therapies for ovarian cancer, bevacizumab, PARP inhibitors and ICIs still fail to meet the therapeutic needs of patients with PROC. ADC-specific delivery payload reduces side effects and has been shown to be superior to conventional monotherapy in PROC, resulting in patient benefits (116). ADCs combined with bevacizumab have displayed superior activity and tolerability in PROC. Therefore, ADCs may become the preferred combination with bevacizumab, providing a superior treatment option for patients with PROC. However, the results from applications in other cancers have shown that ADCs can eventually allow disease progression due to the emergence of drug resistance. These ADC-resistant cells remain sensitive to standard chemotherapeutic agents or other ADCs, which suggests that ADC resistance can be overcome by the appropriate use of other alternative drugs.
Several possible ways to overcome ADC resistance have been proposed, such as the use of cytotoxic agents with poor efflux potential and combinations with other drugs (117,118). With technological advancement, dual-targeting ADCs and dual-drug ADCs have been further developed. Dual-targeting ADCs can recognize and bind to two different targets at the same time and also bind to tumor cells more precisely and effectively, thus overcoming the therapeutic challenges posed by tumor heterogeneity. These strategies are expected to reduce the likelihood of tumor cells becoming resistant to treatment and extend the effective duration of drug therapy (119). Dual-drug ADCs, on the contrary, use two different cytotoxic drugs as payloads; more significant antitumor activity can be obtained, and the occurrence of drug resistance can be reduced by precisely controlling the ratio of the two drugs (120). Enhancing the bystander-killing effect by converting non-cleavable linkers into cleavable linkers is also a way to overcome drug resistance (105). Moreover, ADCs release tumor-associated antigens while killing tumor cells, thus activating the antitumor immune response, making ADCs synergistic with ICIs (121). The combination of the two can enhance the killing effect on tumor cells, thus overcoming the drug resistance of ADCs. For example, one study has shown that combining ADCs and ICIs can improve the ORR of patients, prolong PFS and also have certain therapeutic effects on some patients resistant to ADC in triple-negative breast cancer (NCT03742102). These studies are expected to provide additional insights into the optimal use of these agents.
The optimization of the ADC drug structure and innovative developments are expected to further improve its therapeutic efficacy and safety. Consequent to the ongoing advancement of science and technology, the search for ADCs more precisely targeting tumor cells and more stable in the circulation may represent a prominent research focus in the future. The continuous discovery and validation of new drug targets can propel the advancement of ADCs, thereby broadening the range of ADC applications in clinical settings. This may have a significant and far-reaching impact on the landscape of cancer treatment. Combining ADCs with various antitumor agents is a promising strategy that needs further exploration and optimization to improve efficacy and mitigate adverse effects. The current ADCs in PROC are primarily directed toward high-grade plasmacytoid ovarian cancer. Further studies may include other histological subtypes.
The safety and resistance after long-term drug use needs to be further evaluated as research continues. At present, the research on the discovery, related mechanisms and solutions of drug resistance in ADC mainly focuses on breast cancer, lung cancer and gastric cancer. The corresponding data on ovarian cancer is still lacking. ADC resistance may be discovered in the future due to reduced FRα expression. Therefore, inspiration should be drawn from the studies on other cancers and actively address potential drug resistance by developing novel ADC designs or implementing appropriate alternative and combination therapies.
ADCs have demonstrated potent clinical activity in PROC by selectively delivering cytotoxic drugs to tumor cells, improving therapeutic efficacy and reducing toxicity. The approval of MIRV for treating FRα-positive PROC has led to the rapid evolution of ADC monotherapy and combination therapies, which offers hope for patients with PROC. The gradual use of more effective ADCs for treating PROC may be observed in the future.
The development of novel drugs that can more accurately target tumor cells and are more stable in the circulation will be the future pursuit of researchers. The possible emergence of ADC resistance and the mechanism behind it during clinical application should be considered, so as to more effectively use ADCs as a weapon to optimize the therapeutic regimens for patients.
Not applicable.
Funding: No funding was received.
Not applicable.
KS and SY were responsible for conceptualization, data curation and writing the original draft. NS and FT participated in the topic selection and were responsible for data collection and analysis. SR and HaW were responsible for software and validation, managing the literature and editing the table section of the manuscript. YZ and YW were responsible for creating the figure and participated in drafting the manuscript. HoW and HG were responsible for reviewing and editing the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Matulonis UA, Sood AK, Fallowfield L, Howitt BE, Sehouli J and Karlan BY: Ovarian cancer. Nat Rev Dis Primers. 2:160612016. View Article : Google Scholar : PubMed/NCBI | |
|
St Laurent J and Liu J: Treatment approaches for platinum-resistant ovarian cancer. J Clin Oncol. 42:127–133. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Davis A, Tinker AV and Friedlander M: ‘Platinum resistant’ ovarian cancer: What is it, who to treat and how to measure benefit? Gynecol Oncol. 133:624–631. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Beck A, Goetsch L, Dumontet C and Corvaïa N: Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov. 16:315–337. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Tsuchikama K, Anami Y, Ha SYY and Yamazaki CM: Exploring the next generation of antibody-drug conjugates. Nat Rev Clin Oncol. 21:203–223. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Drago JZ, Modi S and Chandarlapaty S: Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat Rev Clin Oncol. 18:327–344. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Dumontet C, Reichert JM, Senter PD, Lambert JM and Beck A: Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov. 22:641–661. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Gonzalez-Ochoa E, Veneziani AC and Oza AM: Mirvetuximab soravtansine in platinum-resistant ovarian cancer. Clin Med Insights Oncol. 17:117955492311872642023. View Article : Google Scholar : PubMed/NCBI | |
|
Joubert N, Beck A, Dumontet C and Denevault-Sabourin C: Antibody-Drug conjugates: The last decade. Pharmaceuticals (Basel). 13:2452020. View Article : Google Scholar : PubMed/NCBI | |
|
Mathe G, Tran BL and Bernard J: Effect on mouse leukemia 1210 of a combination by diazo-reaction of amethopterin and gamma-globulins from hamsters inoculated with such leukemia by heterografts. C R Hebd Seances Acad Sci. 246:1626–1628. 1958.(In French). PubMed/NCBI | |
|
Perez HL, Cardarelli PM, Deshpande S, Gangwar S, Schroeder GM, Vite GD and Borzilleri RM: Antibody-drug conjugates: Current status and future directions. Drug Discov Today. 19:869–881. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Khongorzul P, Ling CJ, Khan FU, Ihsan AU and Zhang J: Antibody-drug conjugates: A comprehensive review. Mol Cancer Res. 18:3–19. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Samantasinghar A, Sunildutt NP, Ahmed F, Soomro AM, Salih ARC, Parihar P, Memon FH, Kim KH, Kang IS and Choi KH: A comprehensive review of key factors affecting the efficacy of antibody drug conjugate. Biomed Pharmacother. 161:1144082023. View Article : Google Scholar : PubMed/NCBI | |
|
Staudacher AH and Brown MP: Antibody drug conjugates and bystander killing: Is antigen-dependent internalisation required? Br J Cancer. 117:1736–1742. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Dubowchik GM and Walker MA: Receptor-mediated and enzyme-dependent targeting of cytotoxic anticancer drugs. Pharmacol Ther. 83:67–123. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Doronina SO, Toki BE, Torgov MY, Mendelsohn BA, Cerveny CG, Chace DF, DeBlanc RL, Gearing RP, Bovee TD, Siegall CB, et al: Development of potent monoclonal antibody auristatin conjugates for cancer therapy. Nat Biotechnol. 21:778–784. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Lee EK and Liu JF: Antibody-drug conjugates in gynecologic malignancies. Gynecol Oncol. 153:694–702. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Teicher BA and Chari RV: Antibody conjugate therapeutics: Challenges and potential. Clin Cancer Res. 17:6389–6397. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Jain N, Smith SW, Ghone S and Tomczuk B: Current ADC linker chemistry. Pharm Res. 32:3526–3540. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Bargh JD, Isidro-Llobet A, Parker JS and Spring DR: Cleavable linkers in antibody-drug conjugates. Chem Soc Rev. 48:4361–4374. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kovtun YV, Audette CA, Ye Y, Xie H, Ruberti MF, Phinney SJ, Leece BA, Chittenden T, Blättler WA and Goldmacher VS: Antibody-drug conjugates designed to eradicate tumors with homogeneous and heterogeneous expression of the target antigen. Cancer Res. 66:3214–3221. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Tsuchikama K and An Z: Antibody-drug conjugates: Recent advances in conjugation and linker chemistries. Protein Cell. 9:33–46. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Dumontet C and Jordan MA: Microtubule-binding agents: A dynamic field of cancer therapeutics. Nat Rev Drug Discov. 9:790–803. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H, Lin Z, Arnst KE, Miller DD and Li W: Tubulin inhibitor-based antibody-drug conjugates for cancer therapy. Molecules. 22:12812017. View Article : Google Scholar : PubMed/NCBI | |
|
Fuentes-Antrás J, Genta S, Vijenthira A and Siu LL: Antibody-drug conjugates: In search of partners of choice. Trends Cancer. 9:339–354. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Riccardi F, Dal Bo M, Macor P and Toffoli G: A comprehensive overview on antibody-drug conjugates: From the conceptualization to cancer therapy. Front Pharmacol. 14:12740882023. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Z, Li S, Han S, Shi C and Zhang Y: Antibody drug conjugate: The ‘biological missile’ for targeted cancer therapy. Signal Transduct Target Ther. 7:932022. View Article : Google Scholar : PubMed/NCBI | |
|
Kaplon H, Crescioli S, Chenoweth A, Visweswaraiah J and Reichert JM: Antibodies to watch in 2023. MAbs. 15:21534102023. View Article : Google Scholar : PubMed/NCBI | |
|
Yao P, Zhang Y, Zhang S, Wei X, Liu Y, Du C, Hu M, Feng C, Li J, Zhao F, et al: Knowledge atlas of antibody-drug conjugates on CiteSpace and clinical trial visualization analysis. Front Oncol. 12:10398822022. View Article : Google Scholar : PubMed/NCBI | |
|
Luhrs CA and Slomiany BL: A human membrane-associated folate binding protein is anchored by a glycosyl-phosphatidylinositol tail. J Biol Chem. 264:21446–21449. 1989. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Ke J, Zhou XE, Yi W, Brunzelle JS, Li J, Yong EL, Xu HE and Melcher K: Structural basis for molecular recognition of folic acid by folate receptors. Nature. 500:486–489. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao R, Min SH, Wang Y, Campanella E, Low PS and Goldman ID: A role for the proton-coupled folate transporter (PCFT-SLC46A1) in folate receptor-mediated endocytosis. J Biol Chem. 284:4267–4274. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Cheung A, Opzoomer J, Ilieva KM, Gazinska P, Hoffmann RM, Mirza H, Marlow R, Francesch-Domenech E, Fittall M, Rodriguez DD, et al: Anti-Folate receptor alpha-directed antibody therapies restrict the growth of triple-negative breast cancer. Clin Cancer Res. 24:5098–5111. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ledermann JA, Canevari S and Thigpen T: Targeting the folate receptor: Diagnostic and therapeutic approaches to personalize cancer treatments. Ann Oncol. 26:2034–2043. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Heo YA: Mirvetuximab soravtansine: First approval. Drugs. 83:265–273. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Dilawari A, Shah M, Ison G, Gittleman H, Fiero MH, Shah A, Hamed SS, Qiu J, Yu J, Manheng W, et al: FDA approval summary: Mirvetuximab soravtansine-gynx for FRα-positive, platinum-resistant ovarian cancer. Clin Cancer Res. 29:3835–3840. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Oroudjev E, Lopus M, Wilson L, Audette C, Provenzano C, Erickson H, Kovtun Y, Chari R and Jordan MA: Maytansinoid-antibody conjugates induce mitotic arrest by suppressing microtubule dynamic instability. Mol Cancer Ther. 9:2700–2713. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Moore KN, Martin LP, O'Malley DM, Matulonis UA, Konner JA, Vergote I, Ponte JF and Birrer MJ: A review of mirvetuximab soravtansine in the treatment of platinum-resistant ovarian cancer. Future Oncol. 14:123–136. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Tomao F, D'Incalci M, Biagioli E, Peccatori FA and Colombo N: Restoring platinum sensitivity in recurrent ovarian cancer by extending the platinum-free interval: Myth or reality? Cancer. 123:3450–3459. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Ab O, Whiteman KR, Bartle LM, Sun X, Singh R, Tavares D, LaBelle A, Payne G, Lutz RJ, Pinkas J, et al: IMGN853, a folate receptor-α (FRα)-targeting antibody-drug conjugate, exhibits potent targeted antitumor activity against FRα-expressing tumors. Mol Cancer Ther. 14:1605–1613. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Matulonis UA, Lorusso D, Oaknin A, Pignata S, Dean A, Denys H, Colombo N, Van Gorp T, Konner JA, Marin MR, et al: Efficacy and safety of mirvetuximab soravtansine in patients with platinum-resistant ovarian cancer with high folate receptor alpha expression: Results from the SORAYA study. J Clin Oncol. 41:2436–2445. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Martin LP, Konner JA, Moore KN, Seward SM, Matulonis UA, Perez RP, Su Y, Berkenblit A, Ruiz-Soto R and Birrer MJ: Characterization of folate receptor alpha (FRα) expression in archival tumor and biopsy samples from relapsed epithelial ovarian cancer patients: A phase I expansion study of the FRα-targeting antibody-drug conjugate mirvetuximab soravtansine. Gynecol Oncol. 147:402–407. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Moore KN, Angelergues A, Konecny GE, García Y, Banerjee S, Lorusso D, Lee JY, Moroney JW, Colombo N, Roszak A, et al: Mirvetuximab soravtansine in FRα-positive, platinum-resistant ovarian cancer. N Engl J Med. 389:2162–2174. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sidaway P: Mirvetuximab soravtansine superior to chemotherapy in platinum-resistant epithelial ovarian cancer. Nat Rev Clin Oncol. 21:832024. View Article : Google Scholar | |
|
Li X, Zhou S, Abrahams CL, Krimm S, Smith J, Bajjuri K, Stephenson HT, Henningsen R, Hanson J, Heibeck TH, et al: Discovery of STRO-002, a novel homogeneous ADC targeting folate receptor alpha, for the treatment of ovarian and endometrial cancers. Mol Cancer Ther. 22:155–167. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Furuuchi K, Rybinski K, Fulmer J, Moriyama T, Drozdowski B, Soto A, Fernando S, Wilson K, Milinichik A, Dula ML, et al: Antibody-drug conjugate MORAb-202 exhibits long-lasting antitumor efficacy in TNBC PDx models. Cancer Sci. 112:2467–2480. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Shimizu T, Fujiwara Y, Yonemori K, Koyama T, Sato J, Tamura K, Shimomura A, Ikezawa H, Nomoto M, Furuuchi K, et al: First-in-human phase 1 study of MORAb-202, an antibody-drug conjugate comprising farletuzumab linked to eribulin mesylate, in patients with folate receptor-α-positive advanced solid tumors. Clin Cancer Res. 27:3905–3915. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Najjar MK, Manore SG, Regua AT and Lo HW: Antibody-Drug conjugates for the treatment of HER2-positive breast cancer. Genes (Basel). 13:20652022. View Article : Google Scholar : PubMed/NCBI | |
|
Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z and Kurzrock R: HER2 expression status in diverse cancers: Review of results from 37,992 patients. Cancer Metastasis Rev. 34:157–164. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Oh DY and Bang YJ: HER2-targeted therapies-a role beyond breast cancer. Nat Rev Clin Oncol. 17:33–48. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
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. 22:5097–5108. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Mutlu L, McNamara B, Bellone S, Manavella DD, Demirkiran C, Greenman M, Verzosa MSZ, Buza N, Hui P, Hartwich TMP, et al: Trastuzumab deruxtecan (DS-8201a), a HER2-targeting antibody-drug conjugate, demonstrates in vitro and in vivo antitumor activity against primary and metastatic ovarian tumors overexpressing HER2. Clin Exp Metastasis. 41:765–775. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Meric-Bernstam F, Makker V, Oaknin A, Oh DY, Banerjee S, González-Martín A, Jung KH, Ługowska I, Manso L, Manzano A, et al: Efficacy and safety of trastuzumab deruxtecan in patients With HER2-expressing solid tumors: Primary results from the DESTINY-PanTumor02 phase II trial. J Clin Oncol. 42:47–58. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Erickson BK, Zeybek B, Santin AD and Fader AN: Targeting human epidermal growth factor receptor 2 (HER2) in gynecologic malignancies. Curr Opin Obstet Gynecol. 32:57–64. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lipinski M, Parks DR, Rouse RV and Herzenberg LA: Human trophoblast cell-surface antigens defined by monoclonal antibodies. Proc Natl Acad Sci USA. 78:5147–5150. 1981. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Deng J, Yuan Y, Chen W, Sun W, Wang Y, Huang H, Liang B, Ming T, Wen J, et al: Advances in Trop2-targeted therapy: Novel agents and opportunities beyond breast cancer. Pharmacol Ther. 239:1082962022. View Article : Google Scholar : PubMed/NCBI | |
|
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. 20:2329–2340. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
McNamara B, Greenman M, Bellone S, Santin LA, Demirkiran C, Mutlu L, Hartwich TMP, Yang-Hartwich Y, Ratner E, Schwartz PE and Santin AD: Preclinical activity of datopotamab deruxtecan, a novel TROP2 directed antibody-drug conjugate targeting trophoblast cell-surface antigen 2 (TROP2) in ovarian carcinoma. Gynecol Oncol. 189:16–23. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Janjigian YY, Oaknin A, Lang JM, Ciombor KK, Ray-Coquard IL, Oza AM, Yonemori K, Xu RH, Zhao J, Gajavelli S, et al: TROPION-PanTumor03: Phase 2, multicenter study of datopotamab deruxtecan (Dato-DXd) as monotherapy and in combination with anticancer agents in patients (pts) with advanced/metastatic solid tumors. J Clin Oncol. 41:TPS31532023. View Article : Google Scholar | |
|
Cheng WF, Huang CY, Chang MC, Hu YH, Chiang YC, Chen YL, Hsieh CY and Chen CA: High mesothelin correlates with chemoresistance and poor survival in epithelial ovarian carcinoma. Br J Cancer. 100:1144–1153. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Golfier S, Kopitz C, Kahnert A, Heisler I, Schatz CA, Stelte-Ludwig B, Mayer-Bartschmid A, Unterschemmann K, Bruder S, Linden L, et al: Anetumab ravtansine: A novel mesothelin-targeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol Cancer Ther. 13:1537–1548. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Santin AD, Vergote I, González-Martín A, Moore K, Oaknin A, Romero I, Diab S, Copeland LJ, Monk BJ, Coleman RL, et al: Safety and activity of anti-mesothelin antibody-drug conjugate anetumab ravtansine in combination with pegylated-liposomal doxorubicin in platinum-resistant ovarian cancer: Multicenter, phase Ib dose escalation and expansion study. Int J Gynecol Cancer. 33:562–570. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lheureux S, Alqaisi H, Cohn DE, Chern JY, Duska LR, Jewell A, Corr B, Winer IS, Girda E, Crispens MA, et al: A randomized phase II study of bevacizumab and weekly anetumab ravtansine or weekly paclitaxel in platinum-resistant or refractory ovarian cancer NCI trial#10150. J Clin Oncol. 40:55142022. View Article : Google Scholar | |
|
Rottey S, Clarke J, Aung K, Machiels JP, Markman B, Heinhuis KM, Millward M, Lolkema M, Patel SP, de Souza P, et al: Phase I/IIa trial of BMS-986148, an anti-mesothelin antibody-drug conjugate, alone or in combination with nivolumab in patients with advanced solid tumors. Clin Cancer Res. 28:95–105. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Levan K, Mehryar M, Mateoiu C, Albertsson P, Bäck T and Sundfeldt K: Immunohistochemical evaluation of epithelial ovarian carcinomas identifies three different expression patterns of the MX35 antigen, NaPi2b. BMC Cancer. 17:3032017. View Article : Google Scholar : PubMed/NCBI | |
|
Gerber DE, Infante JR, Gordon MS, Goldberg SB, Martín M, Felip E, Garcia MM, Schiller JH, Spigel DR, Cordova J, et al: Phase Ia study of anti-NaPi2b antibody-drug conjugate lifastuzumab vedotin DNIB0600A in patients with non-small cell lung cancer and platinum-resistant ovarian cancer. Clin Cancer Res. 26:364–372. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Moore KN, Birrer MJ, Marsters J, Wang Y, Choi Y, Royer-Joo S, Lemahieu V, Armstrong K, Cordova J, Samineni D, et al: Phase 1b study of anti-NaPi2b antibody-drug conjugate lifastuzumab vedotin (DNIB0600A. in patients with platinum-sensitive recurrent ovarian cancer. Gynecol Oncol. 158:631–639. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Banerjee S, Oza AM, Birrer MJ, Hamilton EP, Hasan J, Leary A, Moore KN, Mackowiak-Matejczyk B, Pikiel J, Ray-Coquard I, et al: Anti-NaPi2b antibody-drug conjugate lifastuzumab vedotin (DNIB0600A) compared with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer in a randomized, open-label, phase II study. Ann Oncol. 29:917–923. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Suzuki H, Nagase S, Saito C, Takatsuka A, Nagata M, Honda K, Kaneda Y, Nishiya Y, Honda T, Ishizaka T, et al: Raludotatug deruxtecan, a CDH6-targeting antibody-drug conjugate with a DNA topoisomerase I inhibitor DXd, is efficacious in human ovarian and kidney cancer models. Mol Cancer Ther. 23:257–271. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gitto SB, Whicker M, Davies G, Kumar S, Kinneer K, Xu H, Lewis A, Mamidi S, Medvedev S, Kim H, et al: A B7-H4-targeting antibody-drug conjugate shows antitumor activity in PARPi and platinum-resistant cancers with B7-H4 expression. Clin Cancer Res. 30:1567–1581. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ponte JF, Ab O, Lanieri L, Lee J, Coccia J, Bartle LM, Themeles M, Zhou Y, Pinkas J and Ruiz-Soto R: Mirvetuximab soravtansine (IMGN853), a folate receptor alpha-targeting antibody-drug conjugate, potentiates the activity of standard of care therapeutics in ovarian cancer models. Neoplasia. 18:775–784. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Quanz M, Hagemann UB, Zitzmann-Kolbe S, Stelte-Ludwig B, Golfier S, Elbi C, Mumberg D, Ziegelbauer K and Schatz CA: Anetumab ravtansine inhibits tumor growth and shows additive effect in combination with targeted agents and chemotherapy in mesothelin-expressing human ovarian cancer models. Oncotarget. 9:34103–34121. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Moore KN, O'Malley DM, Vergote I, Martin LP, Gonzalez-Martin A, Malek K and Birrer MJ: Safety and activity findings from a phase 1b escalation study of mirvetuximab soravtansine, a folate receptor alpha (FRalpha)-targeting antibody-drug conjugate (ADC), in combination with carboplatin in patients with platinum-sensitive ovarian cancer. Gynecol Oncol. 151:46–52. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jain RK: Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science. 307:58–62. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Haunschild CE and Tewari KS: Bevacizumab use in the frontline, maintenance and recurrent settings for ovarian cancer. Future Oncol. 16:225–246. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Pignata S, Lorusso D, Joly F, Gallo C, Colombo N, Sessa C, Bamias A, Salutari V, Selle F, Frezzini S, et al: Carboplatin-based doublet plus bevacizumab beyond progression versus carboplatin-based doublet alone in patients with platinum-sensitive ovarian cancer: A randomised, phase 3 trial. Lancet Oncol. 22:267–276. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
O'Malley DM, Matulonis UA, Birrer MJ, Castro CM, Gilbert L, Vergote I, Martin LP, Mantia-Smaldone GM, Martin AG, Bratos R, et al: Phase Ib study of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC), in combination with bevacizumab in patients with platinum-resistant ovarian cancer. Gynecol Oncol. 157:379–385. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gilbert L, Oaknin A, Matulonis UA, Mantia-Smaldone GM, Lim PC, Castro CM, Provencher D, Memarzadeh S, Method M, Wang J, et al: Safety and efficacy of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC), in combination with bevacizumab in patients with platinum-resistant ovarian cancer. Gynecol Oncol. 170:241–247. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Richardson DL, Moore KN, Vergote I, Gilbert L, Martin LP, Mantia-Smaldone GM, Castro CM, Provencher D, Matulonis UA, Stec J, et al: Phase 1b study of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate, in combination with carboplatin and bevacizumab in patients with platinum-sensitive ovarian cancer. Gynecol Oncol. 185:186–193. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Nicolò E, Giugliano F, Ascione L, Tarantino P, Corti C, Tolaney SM, Cristofanilli M and Curigliano G: Combining antibody-drug conjugates with immunotherapy in solid tumors: current landscape and future perspectives. Cancer Treat Rev. 106:1023952022. View Article : Google Scholar : PubMed/NCBI | |
|
Gerber HP, Sapra P, Loganzo F and May C: Combining antibody-drug conjugates and immune-mediated cancer therapy: What to expect? Biochem Pharmacol. 102:1–6. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Vafa O, Gilliland GL, Brezski RJ, Strake B, Wilkinson T, Lacy ER, Scallon B, Teplyakov A, Malia TJ and Strohl WR: An engineered Fc variant of an IgG eliminates all immune effector functions via structural perturbations. Methods. 65:114–126. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Bauzon M, Drake PM, Barfield RM, Cornali BM, Rupniewski I and Rabuka D: Maytansine-bearing antibody-drug conjugates induce in vitro hallmarks of immunogenic cell death selectively in antigen-positive target cells. Oncoimmunology. 8:e15658592019. View Article : Google Scholar : PubMed/NCBI | |
|
Müller P, Martin K, Theurich S, Schreiner J, Savic S, Terszowski G, Lardinois D, Heinzelmann-Schwarz VA, Schlaak M, Kvasnicka HM, et al: Microtubule-depolymerizing agents used in antibody-drug conjugates induce antitumor immunity by stimulation of dendritic cells. Cancer Immunol Res. 2:741–755. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Iwata TN, Ishii C, Ishida S, Ogitani Y, Wada T and Agatsuma T: A HER2-targeting antibody-drug conjugate, trastuzumab deruxtecan (DS-8201a), enhances antitumor immunity in a mouse model. Mol Cancer Ther. 177:1494–1503. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Junttila TT, Li G, Parsons K, Phillips GL and Sliwkowski MX: Trastuzumab-DM1 (T-DM1. retains all the mechanisms of action of trastuzumab and efficiently inhibits growth of lapatinib insensitive breast cancer. Breast Cancer Res Treat. 128:347–356. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Wu S, Ge A, Deng X, Liu L and Wang Y: Evolving immunotherapeutic solutions for triple-negative breast carcinoma. Cancer Treat Rev. 130:1028172024. View Article : Google Scholar : PubMed/NCBI | |
|
Bardia A, Mayer IA, Diamond JR, Moroose RL, Isakoff SJ, Starodub AN, Shah NC, O'Shaughnessy J, Kalinsky K, Guarino M, et al: Efficacy and safety of anti-trop-2 antibody drug conjugate sacituzumab govitecan (IMMU-132) in heavily pretreated patients with metastatic triple-negative breast cancer. J Clin Oncol. 35:2141–2148. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Camidge DR, Barlesi F, Goldman JW, Morgensztern D, Heist R, Vokes E, Angevin E, Hong DS, Rybkin II, Barve M, et al: A phase 1b study of telisotuzumab vedotin in combination with nivolumab in patients with NSCLC. JTO Clin Res Rep. 3:1002622022.PubMed/NCBI | |
|
Anastasio MK, Shuey S and Davidson BA: Antibody-drug conjugates in gynecologic cancers. Curr Treat Options Oncol. 25:1–19. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Liu L, Jin X and Yu Y: Efficacy and safety of mirvetuximab soravtansine in recurrent ovarian cancer with FRa positive expression: A systematic review and meta-analysis. Crit Rev Oncol Hematol. 194:1042302024. View Article : Google Scholar : PubMed/NCBI | |
|
Modi S, Saura C, Yamashita T, Park YH, Kim SB, Tamura K, Andre F, Iwata H, Ito Y, Tsurutani J, et al: Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med. 382:610–621. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kumagai K, Aida T, Tsuchiya Y, Kishino Y, Kai K and Mori K: Interstitial pneumonitis related to trastuzumab deruxtecan, a human epidermal growth factor receptor 2-targeting Ab-drug conjugate, in monkeys. Cancer Sci. 111:4636–4645. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Swain SM, Nishino M, Lancaster LH, Li BT, Nicholson AG, Bartholmai BJ, Naidoo J, Schumacher-Wulf E, Shitara K, Tsurutani J, et al: Multidisciplinary clinical guidance on trastuzumab deruxtecan (T-DXd)-related interstitial lung disease/pneumonitis-Focus on proactive monitoring, diagnosis, and management. Cancer Treat Rev. 106:1023782022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Y, Liu K, Wang K and Zhu H: Treatment-related adverse events of antibody-drug conjugates in clinical trials: A systematic review and meta-analysis. Cancer. 129:283–295. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Eaton JS, Miller PE, Mannis MJ and Murphy CJ: Ocular adverse events associated with antibody-drug conjugates in human clinical trials. J Ocul Pharmacol Ther. 31:589–604. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Kim SK, Ursell P, Coleman RL, Monk BJ and Vergote I: Mitigation and management strategies for ocular events associated with tisotumab vedotin. Gynecol Oncol. 165:385–392. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kubo K, Azuma A, Kanazawa M, Kameda H, Kusumoto M, Genma A, Saijo Y, Sakai F, Sugiyama Y, Tatsumi K, et al: Consensus statement for the diagnosis and treatment of drug-induced lung injuries. Respir Investig. 51:260–277. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Abelman RO, Wu B, Spring LM, Ellisen LW and Bardia A: Mechanisms of resistance to antibody-drug conjugates. Cancers (Basel). 15:12782023. View Article : Google Scholar : PubMed/NCBI | |
|
Khoury R, Saleh K, Khalife N, Saleh M, Chahine C, Ibrahim R and Lecesne A: Mechanisms of resistance to antibody-drug conjugates. Int J Mol Sci. 24:96742023. View Article : Google Scholar : PubMed/NCBI | |
|
Sung M, Tan X, Lu B, Golas J, Hosselet C, Wang F, Tylaska L, King L, Zhou D, Dushin R, et al: Caveolae-mediated endocytosis as a novel mechanism of resistance to trastuzumab emtansine (T-DM1). Mol Cancer Ther. 17:243–253. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Loganzo F, Sung M and Gerber HP: Mechanisms of resistance to antibody-drug conjugates. Mol Cancer Ther. 15:2825–2834. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Loganzo F, Tan X, Sung M, Jin G, Myers JS, Melamud E, Wang F, Diesl V, Follettie MT, Musto S, et al: Tumor cells chronically treated with a trastuzumab-maytansinoid antibody-drug conjugate develop varied resistance mechanisms but respond to alternate treatments. Mol Cancer Ther. 14:952–963. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Collins DM, Bossenmaier B, Kollmorgen G and Niederfellner G: Acquired resistance to antibody-drug conjugates. Cancers (Basel). 11:3942019. View Article : Google Scholar : PubMed/NCBI | |
|
Yan T, Zhu L and Chen J: Current advances and challenges in CAR T-Cell therapy for solid tumors: tumor-associated antigens and the tumor microenvironment. Exp Hematol Oncol. 12:142023. View Article : Google Scholar : PubMed/NCBI | |
|
Lin L and Lin DC: biological significance of tumor heterogeneity in esophageal squamous cell carcinoma. Cancers (Basel). 11:11562019. View Article : Google Scholar : PubMed/NCBI | |
|
Mitra A, Barua A, Huang L, Ganguly S, Feng Q and He B: From bench to bedside: The history and progress of CAR T cell therapy. Front Immunol. 14:11880492023. View Article : Google Scholar : PubMed/NCBI | |
|
Norberg SM and Hinrichs CS: Engineered T cell therapy for viral and non-viral epithelial cancers. Cancer Cell. 41:58–69. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Paijens ST, Vledder A, de Bruyn M and Nijman HW: Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 18:842–859. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Schlabach MR, Lin S, Collester ZR, Wrocklage C, Shenker S, Calnan C, Xu T, Gannon HS, Williams LJ, Thompson F, et al: Rational design of a SOCS1-edited tumor-infiltrating lymphocyte therapy using CRISPR/Cas9 screens. J Clin Invest. 133:e1630962023. View Article : Google Scholar : PubMed/NCBI | |
|
Muthukutty P and Yoo SY: Oncolytic virus engineering and utilizations: Cancer immunotherapy perspective. Viruses. 15:16452023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang C, Nguyen J and Yen Y: Complete spectrum of adverse events associated with chimeric antigen receptor (CAR)-T cell therapies. J Biomed Sci. 301:892023. View Article : Google Scholar | |
|
Diaby V, Adunlin G, Ali AA, Zeichner SB, de Lima Lopes G, Kohn CG and Montero AJ: Cost-effectiveness analysis of 1st through 3rd line sequential targeted therapy in HER2-positive metastatic breast cancer in the United States. Breast Cancer Res Treat. 160:187–196. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Sato S, Shoji T, Jo A, Otsuka H, Abe M, Tatsuki S, Chiba Y, Takatori E, Kaido Y, Nagasawa T, et al: Antibody-drug conjugates: The new treatment approaches for ovarian cancer. Cancers (Basel). 16:25452024. View Article : Google Scholar : PubMed/NCBI | |
|
García-Alonso S, Ocaña A and Pandiella A: Resistance to antibody-drug conjugates. Cancer Res. 78:2159–2165. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Cabaud O, Berger L, Crompot E, Adélaide J, Finetti P, Garnier S, Guille A, Carbuccia N, Farina A, Agavnian E, et al: Overcoming resistance to anti-nectin-4 antibody-drug Conjugate. Mol Cancer Ther. 21:1227–1235. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Andreev J, Thambi N, Bay AE, Delfino F, Martin J, Kelly MP, Kirshner JR, Rafique A, Kunz A, Nittoli T, et al: Bispecific antibodies and antibody-drug conjugates (ADCs) bridging HER2 and prolactin receptor improve efficacy of HER2 ADCs. Mol Cancer Ther. 16:681–693. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yamazaki CM, Yamaguchi A, Anami Y, Xiong W, Otani Y, Lee J, Ueno NT, Zhang N, An Z and Tsuchikama K: Antibody-drug conjugates with dual payloads for combating breast tumor heterogeneity and drug resistance. Nat Commun. 12:35282021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang AJ, Gao Y, Shi YY, Dai MY and Cai HB: A review of recent advances on single use of antibody-drug conjugates or combination with tumor immunology therapy for gynecologic cancer. Front Pharmacol. 13:10936662022. View Article : Google Scholar : PubMed/NCBI |