Trabectedin (Yondelis®): A Comprehensive Monograph on a Marine-Derived Antineoplastic Agent
Section 1: Executive Summary
Trabectedin, marketed as Yondelis®, represents a significant milestone in oncologic drug development, exemplifying the therapeutic potential harbored within marine ecosystems. Identified by its DrugBank ID DB05109, this small molecule is a complex tetrahydroisoquinoline alkaloid, originally isolated from the Caribbean tunicate Ecteinascidia turbinata and now produced via a scalable semi-synthetic process. Its classification as an alkylating agent belies a unique and multifaceted mechanism of action that distinguishes it from conventional cytotoxic drugs. Trabectedin covalently binds to the minor groove of DNA, inducing a structural distortion that triggers a cascade of events leading to cell cycle arrest and apoptosis. Critically, it interferes with cellular machinery by poisoning the transcription-coupled nucleotide excision repair (TC-NER) pathway and exploiting deficiencies in homologous recombination repair (HRR), providing a strong rationale for its activity in specific tumor types. Furthermore, Trabectedin exerts profound effects on the tumor microenvironment (TME), selectively depleting immunosuppressive tumor-associated macrophages (TAMs).
These distinct mechanisms underpin its approved clinical indications. In Europe, Trabectedin is authorized for the treatment of advanced soft tissue sarcoma (STS) after failure of standard therapies and, in combination with pegylated liposomal doxorubicin (PLD), for relapsed, platinum-sensitive ovarian cancer. In the United States, its approval is more narrowly focused on unresectable or metastatic liposarcoma (LPS) and leiomyosarcoma (LMS) following an anthracycline-containing regimen. The pivotal clinical trials supporting these approvals, ET743-SAR-3007 for sarcoma and OVA-301 for ovarian cancer, demonstrated significant improvements in progression-free survival (PFS), establishing its role in patient populations with limited therapeutic options.
The clinical use of Trabectedin is governed by a significant but manageable safety profile. The most prominent toxicities include neutropenia, which can lead to life-threatening sepsis; hepatotoxicity, characterized by frequent transaminase elevations; and rhabdomyolysis, requiring diligent monitoring of creatine phosphokinase (CPK) levels. Risk mitigation is central to its administration protocol, which mandates the use of a central venous line, a prolonged 24-hour infusion for sarcoma, and obligatory premedication with dexamethasone. This premedication serves a dual purpose, providing both antiemetic control and a crucial hepatoprotective effect. The drug's complex pharmacokinetics, marked by high inter-individual variability and primary metabolism via the CYP3A4 enzyme, necessitate careful management of drug-drug interactions.
Future perspectives for Trabectedin are centered on refining its therapeutic application. Emerging research in pharmacometabolomics offers a potential pathway toward personalized dosing, moving beyond standard body-surface-area calculations to predict individual drug exposure and toxicity. The most promising avenue of investigation lies in immuno-oncology combinations, where Trabectedin’s ability to modulate the TME may synergistically enhance the efficacy of checkpoint inhibitors, as suggested by promising early data from the SAINT trial. Trabectedin thus stands as a testament to the success of natural product drug discovery, a niche but valuable therapeutic agent whose full potential continues to be explored through rational, mechanism-based clinical investigation.
Section 2: Drug Identity, Origin, and Physicochemical Properties
2.1. Identification and Nomenclature
Trabectedin is a well-characterized small molecule antineoplastic agent belonging to the chemical class of alkylating drugs.[1] It is formally registered in major drug and chemical databases under a consistent set of identifiers, ensuring unambiguous recognition in scientific literature, regulatory filings, and clinical practice.
- DrugBank ID: DB05109 [1]
- Type: Small Molecule [1]
- Generic Name: Trabectedin [1]
- Brand Name: Yondelis® [1]
- Synonyms & Developmental Names: During its extensive development, Trabectedin was primarily known by the designation Ecteinascidin 743 or its abbreviation, ET-743. It has also been assigned the National Service Center number NSC 648766.[1]
- Regulatory Identifiers: Key global identifiers include:
- CAS (Chemical Abstracts Service) Number: 114899-77-3 [2]
- European Community (EC) Number: 695-026-8 [2]
- UNII (Unique Ingredient Identifier): ID0YZQ2TCP [2]
2.2. Chemical Structure and Properties
The distinct biological activity of Trabectedin is a direct consequence of its complex and unique molecular architecture. It is a tetrahydroisoquinoline alkaloid characterized by a bridged, polycyclic structure.[2] This intricate three-dimensional conformation, comprising three fused tetrahydroisoquinoline subunits designated as rings A, B, and C, is fundamental to its ability to interact with DNA in a manner distinct from traditional alkylating agents.[9]
- Chemical Formula: The empirical formula for Trabectedin is C39H43N3O11S.[1]
- Molecular Weight: The calculated molecular weight is approximately 761.84 g/mol (average) and 761.261830 g/mol (monoisotopic).[1]
- Physical Properties: In its purified form, Trabectedin is a light yellow to yellow solid.[4] Its physicochemical properties present challenges for formulation, as it demonstrates only slight solubility in common organic solvents such as chloroform and methanol.[4] This necessitates its formulation as a lyophilized powder for reconstitution into a solution for intravenous infusion. The compound has a reported melting point of greater than 143°C, at which point it undergoes decomposition.[4]
2.3. From Marine Tunicate to Synthetic Production: A Developmental Odyssey
The history of Trabectedin is a quintessential narrative of modern natural product drug discovery, illustrating a journey from an obscure marine organism to a clinically vital, synthetically produced medication. This progression was marked by overcoming a critical supply bottleneck that nearly rendered the compound clinically unviable.
The story begins with a broad screening program initiated by the U.S. National Cancer Institute (NCI) in the mid-20th century. In 1969, extracts from the Caribbean marine tunicate, a colonial sea squirt named Ecteinascidia turbinata, were found to possess significant anticancer activity.[3] However, the technology of the era was insufficient to isolate and characterize the active component. It was not until 1984 that Professor K.L. Rinehart at the University of Illinois successfully elucidated the complex structure of the active molecule, which he named Ecteinascidin 743.[3]
The Spanish pharmaceutical company PharmaMar licensed the compound and quickly encountered the primary obstacle to its development: an extremely low natural yield. It was determined that approximately one metric ton (1,000 kilograms) of the tunicate organism was required to isolate a single gram of pure Trabectedin.[3] With several grams needed for even initial clinical trials, harvesting from the wild was unsustainable and commercially impossible. This supply challenge became the central problem to solve.
The first major breakthrough came from the field of synthetic chemistry. At Rinehart's request, the Nobel laureate E.J. Corey and his research group at Harvard University developed and published the first total synthesis of Trabectedin in 1996.[3] This monumental achievement proved that the molecule could be constructed in the laboratory, de-risking the project from its sole reliance on a scarce natural source. While a landmark scientific success, this initial total synthesis was too complex and low-yielding for industrial-scale production.
The pivotal, commercially enabling solution was ultimately developed by PharmaMar. They engineered a more practical and scalable semi-synthetic process. This method begins with a precursor molecule, safracin B, which is produced in large quantities through the fermentation of the bacterium Pseudomonas fluorescens. This precursor is then chemically modified through a series of steps to yield the final Trabectedin molecule.[3] This transition from a rare natural product to a reliable, semi-synthetically manufactured drug was the critical step that unlocked its therapeutic potential, enabling the large-scale clinical trials necessary for regulatory approval and ensuring a stable supply for patients worldwide. This journey serves as a powerful exemplar for the field of pharmacognosy, demonstrating that for natural products, the path to the clinic often depends as much on innovations in chemical and process engineering as on the initial biological discovery.
Table 1: Trabectedin - Key Identifiers and Physicochemical Properties
Identifier Type | Value | Source(s) |
---|
DrugBank ID | DB05109 | 1 |
Generic Name | Trabectedin | 1 |
Brand Name | Yondelis® | 3 |
Developmental Name | Ecteinascidin 743 (ET-743) | 1 |
CAS Number | 114899-77-3 | 2 |
UNII | ID0YZQ2TCP | 2 |
Chemical Formula | C39H43N3O11S | 1 |
Average Molecular Weight | 761.84 g/mol | 7 |
Monoisotopic Molecular Weight | 761.261830 g/mol | 1 |
Physical Form | Solid (Light Yellow to Yellow) | 4 |
Solubility | Slightly soluble in Chloroform, Methanol | 4 |
Chemical Class | Tetrahydroisoquinoline Alkaloid, Alkylating Agent | 2 |
Section 3: Mechanism of Action: A Multi-Pronged Assault on Cancer
Trabectedin exerts its antineoplastic effects through a sophisticated and multi-faceted mechanism of action that is fundamentally different from that of traditional alkylating agents. Its activity is not merely a result of indiscriminate DNA damage but rather a highly specific interaction with the DNA structure, which it then leverages to sabotage critical cellular processes, including transcription and DNA repair. Furthermore, its influence extends beyond the cancer cell itself to modulate the surrounding tumor microenvironment. This combination of direct cytotoxicity and TME remodeling explains its unique spectrum of clinical activity.
3.1. Primary Interaction with DNA: A Minor Groove Specialist
The initial and defining step of Trabectedin's action is its unique binding to DNA. Unlike classic platinum compounds or nitrogen mustards that typically form adducts in the DNA major groove, Trabectedin is a minor groove binder.[1]
The molecule's A and B tetrahydroisoquinoline rings anchor it within the minor groove by forming a reversible covalent bond with the exocyclic N2 amino group of guanine residues.[1] This interaction is not random; Trabectedin exhibits a distinct sequence preference, binding most efficiently at guanine-containing triplets such as 5'-CGG, 5'-TGG, 5'-AGC, or 5'-GGC.[3]
The formation of this Trabectedin-DNA adduct induces a profound structural distortion. The DNA helix is bent by approximately 20 degrees towards the major groove, creating an unusual conformation.[3] This bending is a critical feature of its mechanism. While rings A and B are covalently bound, the third subunit, Ring C, remains unbound and protrudes out from the DNA helix. This protruding portion acts as a physical impediment, sterically hindering the approach and function of nearby nuclear proteins, most notably transcription factors and the enzymes of the DNA repair machinery.[1]
3.2. Sabotage of DNA Repair and Transcription
The true elegance of Trabectedin's mechanism lies in how it converts a cellular defense system into a lethal weapon against the cancer cell. The drug creates a specific type of DNA lesion and then actively prevents the cell's primary repair system from fixing it, a process that culminates in catastrophic and irreparable damage.
- Poisoning the TC-NER Pathway: The structural distortion caused by the Trabectedin adduct is recognized by the Transcription-Coupled Nucleotide Excision Repair (TC-NER) system, a pathway that normally identifies and removes bulky lesions that block RNA polymerase during transcription. However, the interaction with Trabectedin is aberrant. The TC-NER machinery initiates the repair process, making the standard 5' incision in the DNA strand next to the lesion. At this point, the process is sabotaged. The protruding C-ring of the Trabectedin molecule is thought to physically trap the XPG endonuclease, a key enzyme that is supposed to make the second, 3' incision to excise the damaged segment.[9] This failure to complete the repair process results in an abortive cycle, leaving a persistent single-strand break. When the DNA replication fork encounters this stalled and poisoned repair complex, it collapses, generating lethal, irreversible double-strand breaks (DSBs).[3] In this way, the initial lesion is not the primary cytotoxic event; rather, it is the cell's own hijacked repair response that delivers the fatal blow.
- Exploiting Homologous Recombination Deficiency (HRD): The DSBs created by the poisoned TC-NER process are normally repaired by a high-fidelity pathway known as Homologous Recombination Repair (HRR). Many cancers, particularly a subset of ovarian, breast, and prostate cancers, have defects in this pathway, often due to mutations in genes like BRCA1 or BRCA2. This state is known as Homologous Recombination Deficiency (HRD) or "BRCAness." In HRD cells, the DSBs generated by Trabectedin cannot be efficiently repaired. This accumulation of unrepaired DSBs is overwhelmingly toxic, triggering programmed cell death (apoptosis).[18] This provides a powerful molecular rationale for the heightened sensitivity of HRD tumors to Trabectedin and underpins its use in platinum-sensitive ovarian cancer, a disease enriched for this phenotype.
- Inhibition of Gene Transcription: Beyond corrupting DNA repair, the physical presence of the adduct and the distortion of the DNA helix directly interfere with gene transcription. The binding of essential transcription factors to their promoter regions is blocked, leading to the downregulation of specific genes. One of the most significant targets is the multidrug resistance-1 (MDR-1) gene. This gene encodes P-glycoprotein, a membrane-bound efflux pump that actively expels many chemotherapy drugs from the cancer cell, conferring resistance. By inhibiting MDR-1 transcription, Trabectedin may help to overcome or prevent this common mechanism of chemoresistance.[1]
3.3. Modulation of the Tumor Microenvironment (TME)
Trabectedin's antineoplastic activity is not confined to the cancer cell. It also exerts profound and selective effects on the surrounding tumor microenvironment, particularly on the immune cell populations that can either support or suppress tumor growth.
- Selective Depletion of Macrophages: Trabectedin induces rapid and highly selective apoptosis in monocytes and tumor-associated macrophages (TAMs), which are often abundant in tumors and secrete factors that promote proliferation, angiogenesis, and immune evasion.[8] This selective killing is mediated through the activation of caspase-8-dependent apoptosis via the TRAIL (TNF-related apoptosis-inducing ligand) receptors. The selectivity arises because functional TRAIL receptors are highly expressed on monocytes and macrophages, but not on other key leukocyte populations like T-cells or neutrophils, which are therefore spared.[17]
- Inhibition of Pro-inflammatory Milieu: By eliminating TAMs, Trabectedin effectively shuts down a major source of pro-tumorigenic factors. This leads to a marked reduction in the secretion of key cytokines and chemokines, such as C-C Motif Chemokine Ligand 2 (CCL2), Interleukin-6 (IL-6), and Vascular Endothelial Growth Factor (VEGF), within the TME.[9] This action disrupts tumor-promoting inflammation, inhibits angiogenesis, and may help to restore an immune-permissive environment. This TME-modulating effect is so potent that it can contribute to tumor control
in vivo even when the tumor cells themselves are resistant to Trabectedin's direct cytotoxic effects in vitro.[21]
3.4. Specific Activity in Translocation-Related Sarcomas
In certain subtypes of sarcoma defined by specific chromosomal translocations, Trabectedin exhibits an additional, highly targeted mechanism of action. The best-characterized example is in myxoid/round cell liposarcoma, a disease driven by the FUS-CHOP fusion oncoprotein. This aberrant protein acts as a rogue transcription factor, driving a malignant gene expression program.
Trabectedin has been shown to physically displace the FUS-CHOP protein from the promoters of its target genes.[3] This is not a general effect on transcription but a specific interference with the oncogenic driver of the disease. By evicting the fusion protein, Trabectedin reverses the aberrant genetic program, leading to a decrease in proliferation and an increase in adipocytic differentiation, effectively pushing the cancer cells back towards a more normal, non-malignant phenotype.[3] This represents a remarkable example of targeted activity embedded within the drug's broader mechanistic profile and explains its pronounced efficacy in this specific sarcoma subtype.
The clinical activity of Trabectedin can thus be understood as a direct reflection of these distinct mechanisms, each of which is most relevant in a different biological context. Its approval and efficacy in L-sarcomas are driven in large part by its specific activity against oncoproteins like FUS-CHOP. Its approval in platinum-sensitive ovarian cancer is underpinned by the interplay between TC-NER poisoning and the HRD phenotype common in that disease. Finally, its emerging role in combination with immunotherapy is a direct consequence of its potent and selective modulation of the tumor microenvironment. This integrated understanding is essential for appreciating its unique place in therapy and for guiding future clinical development.
Section 4: Clinical Pharmacology
The clinical pharmacology of Trabectedin is complex, characterized by a prolonged exposure profile, high inter-individual variability, and a critical dependence on a single metabolic pathway. These factors collectively define its therapeutic window and dictate the stringent clinical management required for its safe and effective use.
4.1. Pharmacokinetics (Absorption, Distribution, Metabolism, Excretion - ADME)
- Administration and Bioavailability: Trabectedin is formulated for intravenous (IV) administration only, typically as a continuous infusion lasting 3 to 24 hours depending on the indication. As such, its bioavailability is considered 100%.[3] There is no oral formulation.
- Distribution: Following IV administration, Trabectedin exhibits an exceptionally large volume of distribution, reported to be greater than 5,000 liters.[21] This indicates that the drug does not remain confined to the bloodstream but undergoes extensive and rapid distribution into peripheral tissues, where it is sequestered. This extensive tissue binding contributes to its prolonged duration of action.
- Metabolism: Trabectedin undergoes extensive hepatic metabolism. The primary enzyme responsible for its clearance is cytochrome P450 3A4 (CYP3A4).[4] This singular reliance on CYP3A4 is the most critical aspect of its pharmacokinetic profile, as it renders the drug highly susceptible to clinically significant drug-drug interactions.
- Excretion: The drug is characterized by low plasma clearance and a remarkably long terminal elimination half-life, averaging approximately 180 hours (7.5 days).[3] This long half-life, a consequence of its extensive tissue distribution and slow elimination, results in sustained drug exposure long after the infusion is complete.
4.2. Inter-Individual Variability and Pharmacometabolomics
A major challenge in the clinical use of Trabectedin is the high degree of inter-individual variability in its pharmacokinetics. Even with standardized body surface area (BSA)-based dosing, patient exposure can vary significantly, with drug clearance differing by up to 50% between individuals.[26] This variability means that some patients may be under-dosed and derive less benefit, while others may be over-exposed, leading to an increased risk of severe toxicity.
This challenge has spurred innovative research into predictive biomarkers. A landmark pharmacometabolomics study demonstrated that a patient's baseline metabolic profile, measured in plasma before treatment, can explain up to 70% of the observed variability in Trabectedin exposure (as measured by the area under the concentration-time curve, or AUC).[26] The predictive model incorporated five key biomarkers: cystathionine, hemoglobin, taurocholic acid (a primary bile acid), citrulline, and the phenylalanine/tyrosine ratio.
These metabolites are not directly involved in drug metabolism themselves. Instead, they serve as integrated readouts of a patient's underlying physio-pathological state, including hepatic function, systemic inflammation, nutritional status, and tumor burden, all of which can indirectly influence drug disposition.[26] For example, elevated levels of bile acids like taurocholic acid were found to correlate with higher Trabectedin exposure. This may be because bile acids compete with Trabectedin for clearance by CYP3A4 or by hepatic efflux transporters such as ATP-binding cassette C2 (ABCC2, also known as MRP2).[26] This competition could lead to reduced Trabectedin elimination and consequently higher plasma concentrations.
This research represents a potential paradigm shift in the management of Trabectedin therapy. Current practice relies on reactive dose adjustments; a dose is reduced only after a patient has already experienced significant toxicity.[27] Pharmacometabolomics offers a path toward proactive, biomarker-guided personalization. By analyzing a pre-treatment blood sample, clinicians could potentially predict a patient's individual drug clearance capacity. A patient identified as a likely "slow metabolizer" could be started on a lower, safer dose, while a predicted "fast metabolizer" could receive the standard dose with greater confidence. This would be a significant advance over one-size-fits-all BSA dosing, potentially widening the drug's therapeutic index by prospectively minimizing toxicity while preserving efficacy.
4.3. Critical Drug-Drug Interactions
Given its near-total reliance on CYP3A4 for metabolism, Trabectedin is highly susceptible to drug-drug interactions, a fact that is heavily emphasized in its prescribing information and requires vigilant clinical management.
- Strong CYP3A4 Inhibitors: Co-administration with potent inhibitors of the CYP3A4 enzyme must be avoided. These drugs (e.g., azole antifungals like ketoconazole, macrolide antibiotics like clarithromycin, protease inhibitors like ritonavir, and even grapefruit juice) can dramatically slow the metabolism of Trabectedin, leading to a sharp increase in its plasma concentration and a heightened risk of severe or fatal toxicities.[24] Clinical studies have shown that ketoconazole can increase Trabectedin exposure by 66%.[25] If the short-term use of a strong inhibitor is medically necessary, strict guidelines recommend administering the inhibitor at least one week after the Trabectedin infusion and discontinuing it the day before the next infusion.[25]
- Strong CYP3A4 Inducers: Conversely, co-administration with potent inducers of the CYP3A4 enzyme should also be avoided. These agents (e.g., certain anticonvulsants like carbamazepine and phenytoin, rifamycins like rifampin, and the herbal supplement St. John's Wort) can accelerate the metabolism of Trabectedin, leading to a significant decrease in its plasma concentration and a potential loss of therapeutic effect.[1] Studies with rifampin showed a 31% decrease in Trabectedin exposure.[25]
The combination of a long half-life, high intrinsic pharmacokinetic variability, and a narrow therapeutic window makes the safe administration of Trabectedin particularly challenging. The susceptibility to interactions with a wide range of common medications underscores the need for a meticulous review of all concomitant medications by the prescribing oncologist before every treatment cycle.
Section 5: Regulatory History and Approved Clinical Indications
The regulatory journey of Trabectedin has been complex, reflecting the challenges of developing a novel agent for rare diseases. Its path to market varied significantly between major regulatory bodies, particularly concerning its indication in ovarian cancer, providing a compelling case study in differing regulatory philosophies.
5.1. Development and Approval Timeline
The timeline from discovery to clinical use spanned several decades, marked by key scientific and regulatory milestones:
- 1969: Initial discovery of anticancer activity in extracts of the marine tunicate Ecteinascidia turbinata by the NCI.[3]
- 1984: The active compound, Ecteinascidin 743, was structurally elucidated by the laboratory of K.L. Rinehart.[3]
- 1996: First-in-human clinical trials of Trabectedin were initiated, marking its transition into clinical development.[3]
- 2007: The European Medicines Agency (EMA) granted its first major marketing authorization for Yondelis® in the European Union for the treatment of advanced soft tissue sarcoma.[3]
- 2009: The EMA expanded the drug's label to include the treatment of relapsed, platinum-sensitive ovarian cancer when used in combination with pegylated liposomal doxorubicin (PLD).[20]
- 2015: The U.S. Food and Drug Administration (FDA) approved Yondelis® for specific subtypes of advanced soft tissue sarcoma, years after its European approval.[3]
5.2. European Medicines Agency (EMA) Indications
The EMA has approved Trabectedin for two distinct oncologic indications:
- Soft Tissue Sarcoma (STS): Approved in September 2007, Trabectedin is indicated for the treatment of adult patients with advanced STS after failure of first-line chemotherapy with anthracyclines and ifosfamide, or for patients who are unsuited to receive these agents. The EMA noted that the efficacy data were primarily derived from patients with liposarcoma and leiomyosarcoma subtypes.[3] Due to the rarity of the disease and the available data at the time, the initial approval was granted under "exceptional circumstances," a designation that was formally lifted in May 2015 after the submission of additional data.[30]
- Ovarian Cancer: Approved in 2009, Trabectedin is indicated for use in combination with pegylated liposomal doxorubicin (PLD) for the treatment of patients with relapsed, platinum-sensitive ovarian cancer.[20] This approval was based on the results of the pivotal OVA-301 trial. In 2020, the EMA's Committee for Medicinal Products for Human Use (CHMP) conducted a review following the results of a new study (OVC-3006) that did not show benefit in a more platinum-resistant population. The CHMP concluded that the new data were not robust enough to alter the existing benefit-risk profile and reaffirmed the approved indication for platinum-sensitive disease.[33]
5.3. U.S. Food and Drug Administration (FDA) Indications
The FDA's approved indications for Trabectedin are narrower than the EMA's, most notably excluding ovarian cancer.
- Soft Tissue Sarcoma: Yondelis® was approved by the FDA on October 23, 2015. The indication is specifically for the treatment of patients with unresectable or metastatic liposarcoma (LPS) or leiomyosarcoma (LMS) who have previously received an anthracycline-containing chemotherapy regimen.[2]
- Divergence on Ovarian Cancer: The difference in the approved labels for ovarian cancer between the US and EU is a critical aspect of Trabectedin's regulatory history. Following a New Drug Application (NDA) submission in 2008 for the ovarian cancer indication, the FDA issued a Complete Response Letter in 2009 and subsequently requested an additional Phase III study to further support the application.[3] In 2011, the sponsor, Johnson & Johnson, voluntarily withdrew the NDA in the United States.[3] This divergence stems from different interpretations of the available clinical evidence. The pivotal OVA-301 trial demonstrated a statistically significant benefit in the primary endpoint of progression-free survival (PFS).[39] The EMA considered this PFS benefit sufficient for approval in a setting of unmet need. The FDA, however, appeared to place a higher evidentiary bar, likely seeking a more definitive overall survival (OS) benefit, which was not clearly demonstrated in the initial analyses of the trial. The sponsor's decision to withdraw the application rather than conduct another large, costly trial effectively cemented this transatlantic difference in the standard of care for relapsed ovarian cancer, highlighting the ongoing debate in drug regulation regarding the acceptability of surrogate endpoints like PFS for full approval.
5.4. Off-Label and Compendia-Recognized Uses
Despite the relatively narrow FDA-approved label, Trabectedin's utility has been expanded through its inclusion in major clinical practice guidelines, such as those from the National Comprehensive Cancer Network (NCCN). These "off-label" but compendia-recognized uses are considered part of the standard of care in many specialized oncology centers.
These recommendations include the use of single-agent Trabectedin for other STS histologies beyond LPS and LMS, such as angiosarcoma, solitary fibrous tumor, and as a subsequent-line therapy for advanced pleomorphic rhabdomyosarcoma.[41] Furthermore, guidelines support its use in combination with doxorubicin as a first-line or subsequent treatment option for uterine leiomyosarcoma (uLMS), an aggressive subtype of sarcoma.[41] These uses reflect the clinical experience and evidence accumulated outside of the specific context of the registrational trials.
Section 6: Analysis of Pivotal and Ongoing Clinical Trials
The clinical development of Trabectedin has been characterized by a focus on specific, hard-to-treat malignancies. Its approval in both soft tissue sarcoma and ovarian cancer was secured through large Phase III trials that demonstrated a clear, albeit sometimes modest, benefit over existing therapies. Ongoing research continues to explore its potential, particularly in novel combination regimens.
6.1. Soft Tissue Sarcoma (STS)
- Pivotal Trial (ET743-SAR-3007, NCT01343277): This international, randomized, open-label Phase III study was the cornerstone of the FDA approval for Trabectedin in sarcoma. The trial enrolled 518 patients with unresectable or metastatic liposarcoma or leiomyosarcoma who had progressed after receiving an anthracycline-based regimen. Patients were randomized in a 2:1 ratio to receive either Trabectedin (1.5 mg/m² as a 24-hour IV infusion every 3 weeks) or the comparator chemotherapy agent, dacarbazine.[3]
- Key Outcome: The trial successfully met its primary endpoint. Patients treated with Trabectedin demonstrated a statistically significant and clinically meaningful improvement in progression-free survival (PFS) compared to those receiving dacarbazine. The median PFS was 4.2 months in the Trabectedin arm versus only 1.5 months in the dacarbazine arm, corresponding to a 45% reduction in the risk of progression or death (Hazard Ratio = 0.55; p < 0.0001).[14]
- Limitation and Regulatory Context: A key limitation of the study was the lack of a statistically significant difference in the secondary endpoint of overall survival (OS) between the two arms.[14] Despite this, the FDA granted approval based on the robust PFS benefit in this patient population, for whom there were very few effective treatment options available.
- Emerging Combinations (SAINT Trial, NCT03138161): Representing the forefront of Trabectedin research, the SAINT trial is an ongoing Phase 1/2 study evaluating a novel, first-line combination regimen for advanced STS. It combines Trabectedin with two immune checkpoint inhibitors: ipilimumab (an anti-CTLA-4 antibody) and nivolumab (an anti-PD-1 antibody).[45]
- Scientific Rationale: This trial is built on a strong mechanistic hypothesis. Trabectedin's ability to selectively deplete immunosuppressive tumor-associated macrophages (TAMs) from the tumor microenvironment is thought to "prime" the tumor for an immune attack. This remodeling of the TME is expected to synergize with the T-cell activating properties of ipilimumab and nivolumab, leading to a more potent and durable antitumor response.[46]
- Preliminary Results: Early and updated results from the Phase 2 portion of the trial have been highly promising. Across approximately 80-90 evaluable patients, the combination has demonstrated a disease control rate (DCR) of 87.5% and an overall response rate (ORR) of approximately 22-25%, including several complete responses.[46] The reported median OS of 24.6 to 32.0 months appears numerically superior to the historical OS of ~12-18 months seen with standard first-line doxorubicin-based chemotherapy.[46] While the combination carries a significant toxicity profile, it has been deemed manageable, with no unexpected safety signals.[50] This trial suggests a promising future direction for Trabectedin as a key component of immuno-oncology strategies in sarcoma.
6.2. Ovarian Cancer
- Pivotal Trial (OVA-301, NCT00113607): This large, randomized Phase III trial was instrumental in securing the EMA approval for Trabectedin in ovarian cancer. The study enrolled 672 women with relapsed ovarian cancer and compared the combination of Trabectedin (1.1 mg/m² as a 3-hour infusion) plus Pegylated Liposomal Doxorubicin (PLD) against PLD monotherapy.[30]
- Key Outcome: The trial met its primary endpoint, showing that the combination therapy resulted in a statistically significant improvement in PFS compared to PLD alone (median 7.3 months vs. 5.8 months; HR = 0.79; p = 0.0190).[30]
- Subgroup Analysis: A crucial finding from a pre-planned analysis was that the clinical benefit was most pronounced in the subgroup of patients with "partially platinum-sensitive" disease, defined as a platinum-free interval (PFI) of 6 to 12 months. In this cohort, the median PFS was 7.4 months for the combination versus 5.5 months for PLD, and this translated into a significant OS benefit (HR = 0.59).[39] This analysis was critical in defining the optimal patient population for this regimen and shaped its indication in Europe.
- Negative Confirmatory Trial (MITO-23): In contrast to the success of the combination trial, a more recent Phase III study, MITO-23, yielded negative results. This trial evaluated Trabectedin monotherapy against the physician's choice of chemotherapy in a population of patients with recurrent ovarian cancer specifically selected for a high likelihood of sensitivity: those with a germline BRCA1/2 mutation or a "BRCAness" phenotype (i.e., repeated responses to platinum).[22]
- Key Outcome: The trial failed to meet its primary endpoint. Trabectedin monotherapy did not improve OS compared to the control arm (median 15.8 months vs. 17.9 months) and was associated with a worse safety profile.[22] It showed no superiority, even in this theoretically hypersensitive population.
The collective evidence from these trials paints a clear picture of a drug whose efficacy is highly context-dependent. In sarcoma, it established a foothold against a weak comparator and is now being repositioned as a promising partner for immunotherapy. In ovarian cancer, the data strongly suggest that its clinical benefit is realized primarily when used in combination with PLD and within a specific subset of platinum-sensitive patients. The failure of the MITO-23 trial tempers enthusiasm for its use as a single agent, even in biologically selected populations, highlighting that the preclinical rationale of HRD sensitivity does not automatically translate to clinical superiority without the right combination partner.
Table 2: Summary of Pivotal Phase III Clinical Trials
Trial ID | Indication | Treatment Arms | N | Primary Endpoint | Median PFS (Exp. vs. Ctrl.) | HR (PFS) | Median OS (Exp. vs. Ctrl.) | HR (OS) | Key Conclusion |
---|
ET743-SAR-3007 | Advanced Liposarcoma / Leiomyosarcoma | Trabectedin vs. Dacarbazine | 518 | Progression-Free Survival (PFS) | 4.2 vs. 1.5 months | 0.55 | Not Significantly Different | N/A | Trabectedin significantly improves PFS vs. dacarbazine, leading to FDA approval. 14 |
OVA-301 | Relapsed Ovarian Cancer | Trabectedin + PLD vs. PLD Alone | 672 | Progression-Free Survival (PFS) | 7.3 vs. 5.8 months | 0.79 | 22.4 vs. 18.9 months | 0.85 | Combination therapy significantly improves PFS vs. PLD alone, leading to EMA approval. 30 |
Section 7: Safety, Tolerability, and Risk Management
The clinical application of Trabectedin is intrinsically linked to the management of its distinct and significant toxicity profile. While many adverse effects are manageable with supportive care, several serious risks necessitate a rigorous protocol of monitoring, premedication, and dose modification.
7.1. Comprehensive Adverse Event Profile
- Most Common Adverse Reactions: The most frequently reported adverse events (occurring in ≥20% of patients) are largely constitutional and gastrointestinal. These include nausea (75%), fatigue (69%), vomiting (46%), constipation (37%), decreased appetite (37%), diarrhea (35%), peripheral edema (28%), dyspnea (25%), and headache (25%).[3] While common, these are typically Grade 1-2 in severity and can be managed with standard antiemetics and supportive measures.
- Common Grade 3-4 Laboratory Abnormalities: The most significant impact of Trabectedin is seen on laboratory parameters. Severe (Grade 3 or 4) abnormalities are common and form the basis for dose-limiting toxicity. These include neutropenia (43%), elevated alanine aminotransferase (ALT) (31%), thrombocytopenia (21%), anemia (19%), and elevated aspartate aminotransferase (AST) (17%).[27]
7.2. Serious Risks and Management (Boxed Warning-Level Concerns)
The prescribing information for Trabectedin highlights several potentially life-threatening toxicities that require specific risk management strategies.
- Neutropenic Sepsis: Severe neutropenia is the most common dose-limiting toxicity. In the pivotal sarcoma trial, 43% of patients developed Grade 3/4 neutropenia, with a median onset of 16 days. This can lead to febrile neutropenia (in ~5% of patients) and neutropenic sepsis, which has been fatal in approximately 1% of cases.[27] Management requires strict monitoring of absolute neutrophil count (ANC) prior to each treatment cycle and withholding the dose for an ANC below 1,500 cells/µL. Dose reduction is mandated for episodes of febrile neutropenia or prolonged severe neutropenia.[34]
- Rhabdomyolysis: Trabectedin can cause severe muscle toxicity, leading to rhabdomyolysis. This is characterized by marked elevations in creatine phosphokinase (CPK), myalgia, and can progress to myoglobinuria and acute renal failure. Fatal cases of rhabdomyolysis have been reported (in ~0.8% of patients).[34] Monitoring of CPK levels prior to each administration is mandatory. The drug must be withheld for CPK levels more than 2.5 times the upper limit of normal (ULN) and permanently discontinued if rhabdomyolysis is confirmed.[24]
- Hepatotoxicity: Liver toxicity is a very common and serious adverse effect. Grade 3/4 elevations in liver transaminases (AST and ALT) occur in over 30% of patients, and severe, sometimes fatal, hepatic failure can occur.[4] Liver function tests (bilirubin, AST, ALT) must be closely monitored before every cycle. The mandatory premedication with dexamethasone is a critical component of risk mitigation. Beyond its antiemetic properties, dexamethasone provides an essential hepatoprotective benefit.[4] This is thought to be mediated through the induction of hepatic drug transporters, such as ABCC2, which facilitate the clearance of Trabectedin and its metabolites, thereby reducing their toxic accumulation in the liver.[26] This represents a sophisticated pharmacological strategy embedded directly into the administration protocol to enhance the drug's safety.
- Cardiomyopathy: The drug can induce cardiomyopathy, including decreases in left ventricular ejection fraction (LVEF), congestive heart failure, and diastolic dysfunction. In clinical trials, cardiomyopathy occurred in 6% of patients, with some cases being severe (Grade 3/4).[19] Therefore, assessment of LVEF by echocardiogram (ECHO) or a multigated acquisition (MUGA) scan is required at baseline and at 2- to 3-month intervals throughout treatment.[19]
- Extravasation: Trabectedin is a potent vesicant. If the drug leaks outside of the vein during infusion (extravasation), it can cause severe and progressive tissue necrosis, often requiring surgical debridement.[19] To mitigate this serious risk, administration of Trabectedin exclusively through a secure central venous line is mandatory.[19]
- Embryo-Fetal Toxicity: Based on its mechanism of action and findings in animal studies, Trabectedin can cause fetal harm when administered to a pregnant woman. It is designated as Pregnancy Category D in Australia.[3] Effective contraception is required for both male and female patients of reproductive potential during and for several months after treatment completion.
7.3. Contraindications and Special Populations
- Contraindications: The primary contraindication is a history of known severe hypersensitivity, including anaphylaxis, to Trabectedin or any of its components.[24] Concomitant administration with the yellow fever vaccine is also contraindicated due to the risk of fatal systemic disease.[4]
- Hepatic Impairment: Given its extensive hepatic metabolism, liver function is a critical determinant of safety. A specific dose reduction (from 1.5 mg/m² to 0.9 mg/m²) is required for patients with moderate hepatic impairment (bilirubin >1.5 to 3 times ULN). The drug is contraindicated and should not be administered to patients with severe hepatic impairment (bilirubin >3 times ULN).[24]
- Renal Impairment: No dose adjustment is required for patients with mild to moderate renal impairment (CrCl 30-89 mL/min). However, Trabectedin has not been studied in patients with severe renal impairment (CrCl <30 mL/min) or end-stage renal disease, and its use in this population is not recommended.[25]
Section 8: Dosing, Administration, and Clinical Management
The clinical use of Trabectedin is governed by a highly structured and demanding administration protocol designed to maximize its therapeutic index while mitigating its significant toxicities. This protocol inherently restricts its use to specialized oncology centers equipped with the necessary infrastructure and expertise to manage its complexities.
8.1. Prescribing Information and Dosing
The recommended dosage and schedule for Trabectedin differ based on the approved indication and regulatory jurisdiction.
- Standard Dose (Soft Tissue Sarcoma - U.S. and EU): For the treatment of liposarcoma and leiomyosarcoma, the standard dose is 1.5 mg/m² administered as a continuous intravenous infusion over 24 hours. This is repeated every 21 days (3 weeks) until disease progression or unacceptable toxicity occurs.[24]
- Standard Dose (Ovarian Cancer - EU only): For the treatment of relapsed, platinum-sensitive ovarian cancer, Trabectedin is given at a dose of 1.1 mg/m² as a shorter 3-hour intravenous infusion. It is administered every 21 days, immediately following the infusion of its combination partner, pegylated liposomal doxorubicin (PLD).[30]
- Administration Requirements: Due to the severe risk of tissue necrosis from extravasation, Trabectedin must be administered through a central venous line (e.g., a PICC line or port-a-cath). The use of a peripheral IV line is contraindicated. The infusion set must also include a 0.2-micron polyethersulfone (PES) in-line filter.[24]
The logistical burden of these requirements is substantial. A 24-hour infusion necessitates either an inpatient hospital admission or a highly sophisticated ambulatory infusion program with portable pumps and 24-hour on-call support. The requirement for a central line involves an invasive procedure and specialized nursing care. This complex protocol effectively concentrates the use of Trabectedin in major academic medical centers and large cancer institutions with the resources to manage it safely.
8.2. Premedication and Monitoring
A strict regimen of premedication and laboratory monitoring is mandatory for every treatment cycle to manage expected toxicities.
- Mandatory Premedication: All patients must receive dexamethasone 20 mg intravenously approximately 30 minutes prior to each Trabectedin infusion. As previously discussed, this serves the dual purpose of preventing nausea and vomiting and providing critical hepatoprotection.[4]
- Required Monitoring: Before the administration of each cycle, a specific panel of laboratory tests must be performed and reviewed to ensure the patient has recovered from the previous cycle's toxicities. This includes:
- Hematology: Complete blood count with differential to assess absolute neutrophil count (ANC) and platelet count.[24]
- Liver Function: Bilirubin, AST, ALT, and alkaline phosphatase (ALP).[24]
- Muscle Toxicity: Creatine phosphokinase (CPK).[24]
- Cardiac Monitoring: Left Ventricular Ejection Fraction (LVEF) must be assessed by ECHO or MUGA scan at baseline before starting therapy and then periodically every 2 to 3 months throughout the course of treatment to monitor for potential cardiomyopathy.[19]
8.3. Dose Modification and Discontinuation Criteria
The prescribing information provides explicit, data-driven rules for managing toxicities through dose adjustments. These rules are designed to ensure patient safety while attempting to maintain therapeutic benefit.
Doses may be delayed for up to 3 weeks to allow for recovery from toxicity. If recovery takes longer than 3 weeks, the drug should be permanently discontinued. Dose reductions are tiered (e.g., for the sarcoma regimen, from 1.5 mg/m² to 1.2 mg/m², then to 1.0 mg/m²). Once a dose has been reduced due to toxicity, it should not be re-escalated in subsequent cycles.[27] Permanent discontinuation is mandated for a number of severe or persistent adverse events, including rhabdomyolysis, symptomatic cardiomyopathy, or adverse reactions that require dose reduction below the final tier.[27]
Table 3: Dose Modification Guidelines for Adverse Reactions (Sarcoma Regimen)
Laboratory Parameter or Adverse Reaction | Delay Next Dose Until Recovery If: | Reduce Next Dose by 1 Level If During Prior Cycle: |
---|
Platelets | < 100,000/µL | < 25,000/µL |
Absolute Neutrophil Count (ANC) | < 1,500/µL | < 1,000/µL with fever/infection, OR < 500/µL lasting > 5 days |
Total Bilirubin | > Upper Limit of Normal (ULN) | > ULN (requiring dose delay) |
AST or ALT | > 2.5 x ULN | > 5 x ULN |
Alkaline Phosphatase (ALP) | > 2.5 x ULN | > 2.5 x ULN (requiring dose delay) |
Creatine Phosphokinase (CPK) | > 2.5 x ULN | > 5 x ULN |
Other Non-Hematologic Reactions | Grade 3 or 4 | Grade 3 or 4 (requiring dose delay) |
Source: Adapted from Yondelis® Prescribing Information [27]
Section 9: Conclusion and Future Perspectives
Trabectedin (Yondelis®) has firmly established itself as a unique and valuable, albeit niche, therapeutic agent in the modern oncology armamentarium. Its journey from the marine tunicate Ecteinascidia turbinata to a semi-synthetically produced drug is a triumph of natural product development, overcoming formidable supply challenges to deliver a novel mechanism of action to the clinic. Its intricate, multi-pronged assault on cancer—involving minor groove DNA binding, sabotage of the TC-NER pathway, exploitation of HRD, and potent modulation of the tumor microenvironment—sets it apart from all other cytotoxic agents. This complex biology underpins its proven efficacy in specific clinical contexts: as a second-line therapy for advanced lipo- and leiomyosarcomas, and, in Europe, as a combination therapy with PLD for relapsed, platinum-sensitive ovarian cancer.
The clinical use of Trabectedin is demanding, defined by a significant toxicity profile that requires rigorous monitoring, mandatory premedication, and strict adherence to administration protocols. These challenges have concentrated its use in specialized centers but have not diminished its importance for patients with limited alternative options.
Looking forward, the evolution of Trabectedin is likely to proceed along three key strategic paths, moving it from a niche cytotoxic agent toward a more refined, mechanism-driven therapeutic tool:
- The Pursuit of Biomarker-Driven Therapy: The high inter-individual pharmacokinetic variability of Trabectedin is a major clinical challenge. The pioneering research in pharmacometabolomics offers a tantalizing glimpse into a future of personalized dosing. Moving beyond BSA-based calculations to use pre-treatment metabolic signatures to predict a patient's drug exposure could allow for proactive dose adjustments, potentially minimizing severe toxicity and widening the drug's therapeutic window. Further validation of biomarkers related to its mechanism, such as HRD status, may also help refine patient selection for specific indications.
- The Synergy of Immuno-Oncology Combinations: Perhaps the most exciting frontier for Trabectedin is its integration into immuno-oncology regimens. The strong scientific rationale—using Trabectedin to deplete immunosuppressive TAMs and remodel the TME to be more receptive to an immune response—is already bearing fruit. The promising early results of the SAINT trial, combining Trabectedin with dual checkpoint blockade in first-line sarcoma, suggest that this strategy could significantly improve outcomes and potentially reposition the drug as a cornerstone of combination immunotherapy in select solid tumors.
- Exploration of New Mechanistic Niches: The unique sensitivity of HRD-deficient cells to Trabectedin provides a compelling reason to continue exploring its activity in other malignancies known to harbor this phenotype, such as certain subsets of breast, prostate, and pancreatic cancer. While monotherapy has proven challenging, rational combinations designed to exploit this vulnerability could unlock new therapeutic applications.
In conclusion, Trabectedin is more than just another chemotherapy drug. It is a sophisticated biological probe and a therapeutic agent whose complex mechanisms are still being fully elucidated. While it will likely remain a drug for specific situations, its future lies in leveraging its unique biological properties with greater precision. Through biomarker-guided personalization and rational, synergistic combinations, Trabectedin is poised to continue making meaningful contributions to the treatment of cancer for years to come.
Works cited
- Trabectedin: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed July 25, 2025, https://go.drugbank.com/drugs/DB05109
- Trabectedin | C39H43N3O11S | CID 108150 - PubChem, accessed July 25, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Trabectedin
- Trabectedin - Wikipedia, accessed July 25, 2025, https://en.wikipedia.org/wiki/Trabectedin
- Trabectedin | 114899-77-3 - ChemicalBook, accessed July 25, 2025, https://www.chemicalbook.com/ChemicalProductProperty_IN_CB31364731.htm
- Trabectedin (Ecteinascidin 743, ET-743, NSC 648766, CAS Number: 114899-77-3), accessed July 25, 2025, https://www.caymanchem.com/product/34662/trabectedin
- Trabectedin | Apoptosis | Reactive Oxygen Species - TargetMol, accessed July 25, 2025, https://www.targetmol.com/compound/trabectedin
- Trabectedin ≥98% (HPLC) - Sigma-Aldrich, accessed July 25, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/sml4074
- Trabectedin: A review of its use in the management of soft tissue sarcoma and ovarian cancer - ResearchGate, accessed July 25, 2025, https://www.researchgate.net/publication/5918502_Trabectedin_A_review_of_its_use_in_the_management_of_soft_tissue_sarcoma_and_ovarian_cancer
- Mechanism of Action SUMMARY • Trabectedin is an alkylating agent ..., accessed July 25, 2025, https://www.jnjmedicalconnect.com/media/attestation/products/yondelis/yondelis-mechanism-of-action.pdf
- Trabectedin - PMC, accessed July 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2766471/
- pmc.ncbi.nlm.nih.gov, accessed July 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2766471/#:~:text=Trabectedin%20is%20an%20alkylating%20cytostatic,C%20interacts%20with%20transcription%20factors.
- Trabectedin | C39H43N3O11S - ChemSpider, accessed July 25, 2025, https://www.chemspider.com/Chemical-Structure.97236.html
- Trabectedin | 99.92%(HPLC) | In Stock - Selleck Chemicals, accessed July 25, 2025, https://www.selleckchem.com/products/trabectedin.html
- FDA Approves Trabectedin for Sarcoma - NCI, accessed July 25, 2025, https://www.cancer.gov/news-events/cancer-currents-blog/2015/fda-trabectedin-sarcoma
- From Sea to Lab - ChemistryViews, accessed July 25, 2025, https://www.chemistryviews.org/details/ezine/11136900/From_Sea_to_Lab/
- www.yondelis.com, accessed July 25, 2025, https://www.yondelis.com/hcp/mechanism-of-action.html#:~:text=How%20YONDELIS%C2%AE%20works,the%20minor%20groove%20of%20DNA.&text=This%20forms%20adducts%20that%20bend%20the%20DNA%20molecule%20toward%20the%20major%20groove.&text=Adducts%20can%20affect%20the%20activity%20of%20transcription%20factors%20and%20DNA%20repair%20mechanisms.
- Exploiting divergent mechanisms of trabectedin for bone tumors - PMC, accessed July 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11937656/
- Focus on Trabectedin in Ovarian Cancer: What Do We Still Need to Know? - PMC, accessed July 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11166157/
- Mechanism of Action - YONDELIS®, accessed July 25, 2025, https://www.yondelis.com/hcp/mechanism-of-action.html
- Clinical utility of trabectedin for the treatment of ovarian cancer: current evidence - PMC, accessed July 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4103925/
- Trabectedin and lurbinectedin: Mechanisms of action, clinical impact, and future perspectives in uterine and soft tissue sarcoma, ovarian carcinoma, and endometrial carcinoma, accessed July 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9671549/
- Single-Agent Trabectedin Versus Physician's Choice Chemotherapy in Patients With Recurrent Ovarian Cancer With BRCA-Mutated and/or BRCAness Phenotype: A Randomized Phase III Trial - ASCO Publications, accessed July 25, 2025, https://ascopubs.org/doi/10.1200/JCO.23.01225
- Focus on Trabectedin in Ovarian Cancer: What Do We Still Need to Know? | DDDT, accessed July 25, 2025, https://www.dovepress.com/focus-on-trabectedin-in-ovarian-cancer-what-do-we-still-need-to-know-peer-reviewed-fulltext-article-DDDT
- Trabectedin Dosing and Administration | YONDELIS®, accessed July 25, 2025, https://www.yondelis.com/hcp/dosing-admin.html
- Yondelis (trabectedin) dosing, indications, interactions, adverse ..., accessed July 25, 2025, https://reference.medscape.com/drug/yondelis-trabectedin-999997
- Pharmacometabolomics of trabectedin in metastatic soft ... - Frontiers, accessed July 25, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2023.1212634/full
- YONDELIS® - Dosing and Administration Guide, accessed July 25, 2025, https://www.yondelis.com/assets/files/RE-REVIEW_W-CHANGES_YONDELIS_Dosing_and_Administration_Guide.pdf
- Yondelis (trabectedin, ET-743): the development of an anticancer agent of marine origin - PubMed, accessed July 25, 2025, https://pubmed.ncbi.nlm.nih.gov/12960733/
- Trabectedin for the treatment of advanced soft tissue sarcoma | Guidance - NICE, accessed July 25, 2025, https://www.nice.org.uk/Guidance/ta185
- Yondelis | European Medicines Agency (EMA), accessed July 25, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/yondelis
- Yondelis (trabectedin) FDA Approval History - Drugs.com, accessed July 25, 2025, https://www.drugs.com/history/yondelis.html
- Public Assessment Report - Geneesmiddeleninformatiebank, accessed July 25, 2025, https://www.geneesmiddeleninformatiebank.nl/pars/h127706.pdf
- Yondelis - referral | European Medicines Agency (EMA), accessed July 25, 2025, https://www.ema.europa.eu/en/medicines/human/referrals/yondelis
- U.S. FDA Approves YONDELIS® (trabectedin) for the Treatment of ..., accessed July 25, 2025, https://www.jnj.com/media-center/press-releases/us-fda-approves-yondelis-trabectedin-for-the-treatment-of-patients-with-unresectable-or-metastatic-liposarcoma-or-leiomyosarcoma-two-common-subtypes-of-soft-tissue-sarcoma
- FDA Approves Trabectedin (Yondelis) for Advanced Soft-Tissue Sarcoma - CancerNetwork, accessed July 25, 2025, https://www.cancernetwork.com/view/fda-approves-trabectedin-yondelis-advanced-soft-tissue-sarcoma
- Trabectedin Now FDA-Approved for Metastatic Soft Tissue Sarcomas - Oncology Nursing News, accessed July 25, 2025, https://www.oncnursingnews.com/view/trabectedin-now-fda-approved-for-metastatic-soft-tissue-sarcoma
- Clinical Policy: Trabectedin (Yondelis) - Reference Number: LA.PHAR.204 Effective Date - Louisiana Department of Health, accessed July 25, 2025, https://ldh.la.gov/assets/medicaid/PharmPC/9.15.22/LA.PHAR.204TrabectedinYondelis.pdf
- Clinical Policy: Trabectedin (Yondelis) - Louisiana Department of Health, accessed July 25, 2025, https://ldh.la.gov/assets/medicaid/PharmPC/8.5.21/2/LA.PHAR.204TrabectedinYondelis.pdf
- Trabectedin plus pegylated liposomal doxorubicin in relapsed ovarian cancer: outcomes in the partially platinum-sensitive (platinum-free interval 6–12 months) subpopulation of OVA-301 phase III randomized trial, accessed July 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3003616/
- ESGO: Role for Trabectedin Plus Pegylated Doxorubicin in Advanced Ovarian Cancer, OVA-301 Final Results Presented at ESGO - CancerNetwork, accessed July 25, 2025, https://www.cancernetwork.com/view/esgo-role-trabectedin-plus-pegylated-doxorubicin-advanced-ovarian-cancer-ova-301-final-results
- Trabectedin (Yondelis) - Medical Clinical Policy Bulletins | Aetna, accessed July 25, 2025, https://www.aetna.com/cpb/medical/data/900_999/0903.html
- Clinical Policy: Trabectedin (Yondelis) - PA Health & Wellness, accessed July 25, 2025, https://www.pahealthwellness.com/content/dam/centene/Pennsylvania/policies/clinical-policies/Trabectedin-Yondelis-v1.2024.pdf
- Clinical Policy: Trabectedin (Yondelis) - Ambetter Health, accessed July 25, 2025, https://www.ambetterhealth.com/content/dam/centene/ar-health-wellness/policies/Ambetter/Ambetter_Pharmacy_Policy/AR_PHAR_204_Trabectedin.pdf
- A Study of Trabectedin or Dacarbazine for the Treatment of Patients With Advanced Liposarcoma or Leiomyosarcoma | ClinicalTrials.gov, accessed July 25, 2025, https://clinicaltrials.gov/study/NCT01343277
- Study Details | SAINT:Trabectedin, Ipilimumab and Nivolumab for ..., accessed July 25, 2025, https://clinicaltrials.gov/study/NCT03138161
- SAINT: A Phase I/Expanded Phase II Study Using Safe Amounts of Ipilimumab, Nivolumab and Trabectedin as First-Line Treatment of Advanced Soft Tissue Sarcoma, accessed July 25, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9913367/
- First-Line Combo Yields Strong Results in Advanced Sarcoma, accessed July 25, 2025, https://www.ajmc.com/view/first-line-combo-yields-strong-results-in-advanced-sarcoma
- SAINT: Results of an expanded phase II study using safe amounts of ipilimumab (I), nivolumab (N), and trabectedin (T) as first-line treatment of advanced soft tissue sarcoma [NCT03138161]. | Journal of Clinical Oncology - ASCO Publications, accessed July 25, 2025, https://ascopubs.org/doi/10.1200/JCO.2020.38.15_suppl.11520
- Study Details | SAINT:Trabectedin, Ipilimumab and Nivolumab for Previously Treated Advanced Soft Tissue Sarcoma | ClinicalTrials.gov, accessed July 25, 2025, https://clinicaltrials.gov/study/NCT03138161?term=NCT00102609%20NCT01303094%20NCT01710176%20NCT02672527%20NCT02805725%20NCT03138161%20NCT03590210%20NCT03838744%20NCT05597917&rank=4
- Five-year results of a phase 2 trial using ipilimumab (I), nivolumab (N), and trabectedin (T) for previously untreated advanced soft tissue sarcoma (NCT03138161). - ResearchGate, accessed July 25, 2025, https://www.researchgate.net/publication/367695525_Five-year_results_of_a_phase_2_trial_using_ipilimumab_I_nivolumab_N_and_trabectedin_T_for_previously_untreated_advanced_soft_tissue_sarcoma_NCT03138161
- Five-year results of a phase 2 trial using ipilimumab (I), nivolumab (N), and trabectedin (T) for previously untreated advanced soft tissue sarcoma (NCT03138161). - ASCO Publications, accessed July 25, 2025, https://ascopubs.org/doi/10.1200/JCO.2022.40.16_suppl.11573
- Study Details | An Efficacy and Safety Study for Yondelis ..., accessed July 25, 2025, https://clinicaltrials.gov/study/NCT00113607
- Trabectedin plus pegylated liposomal doxorubicin (PLD) prior to subsequent platinum chemotherapy in patients with platinum-resistant (PR) recurrent ovarian cancer (ROC): Results from OVA-301 follow-up. - ASCO Publications, accessed July 25, 2025, https://ascopubs.org/doi/10.1200/jco.2014.32.15_suppl.5551
- Trabectedin plus pegylated liposomal doxorubicin in relapsed ovarian cancer: outcomes in the partially platinum-sensitive (platinum-free interval 6-12 months) subpopulation of OVA-301 phase III randomized trial - PubMed, accessed July 25, 2025, https://pubmed.ncbi.nlm.nih.gov/20643862/
- Trabectedin - ZenOnco.io, accessed July 25, 2025, https://zenonco.io/cancer/trabectedin/
- A Study of Trabectedin in Patients With Advanced Ovarian Cancer - ClinicalTrials.gov, accessed July 25, 2025, https://clinicaltrials.gov/study/NCT00050414