C6H12N3PS
52-24-4
Adenocarcinoma of the Ovaries, Breast Adenocarcinoma, Papillary transitional cell carcinoma of bladder, Malignant effusion
Thiotepa is a seminal organophosphorus alkylating agent with a rich and evolving clinical history that spans more than six decades.[1] Originally synthesized in the early 1950s for applications in the textile industry, its potent effects on rapidly dividing cells were quickly recognized, leading to its investigation as a chemotherapeutic agent.[3] The drug's core antineoplastic mechanism is rooted in its ability to form highly reactive ethylenimine radicals that induce covalent cross-links within the DNA double helix, a non-cell-cycle-specific action that disrupts cellular replication and triggers apoptosis.[1] This fundamental mechanism underpins its broad-spectrum activity against a variety of malignancies.
In the landscape of modern oncology, Thiotepa holds a dual identity. It maintains its historical role in the palliative treatment of several solid tumors, including adenocarcinoma of the breast and ovary, and superficial papillary carcinoma of the bladder.[1] However, its most critical contemporary application is as a high-dose conditioning agent prior to hematopoietic stem cell transplantation (HSCT). In this setting, its potent myelosuppressive and immunosuppressive properties are leveraged to eradicate residual malignancy and facilitate engraftment, making it a cornerstone of treatment for numerous hematologic and central nervous system (CNS) cancers.[1]
The clinical utility of Thiotepa is inextricably linked to its significant toxicity profile, which is prominently highlighted by U.S. Food and Drug Administration (FDA) Boxed Warnings for severe myelosuppression and carcinogenicity.[10] These profound risks dictate its therapeutic index and necessitate meticulous patient monitoring and management. This report provides a definitive, integrated analysis of Thiotepa's pharmacology, clinical efficacy across its historical and modern indications, evolving regulatory landscape, and comprehensive risk management profile, synthesizing decades of data with contemporary clinical evidence to create an exhaustive monograph on this enduring therapeutic agent.
A precise understanding of Thiotepa's identity is fundamental, particularly for a compound with a long history that has resulted in numerous synonyms, code names, and brand names. Thiotepa is a white, crystalline solid, which can present as fine white flakes when crystallized from pentane or ether.[13] Its melting point is reported to be in the range of 51.5°C to 57°C.[13] The compound is soluble in water, with a solubility of 19 g per 100 mL at 25°C, and is also soluble in common organic solvents such as ethanol, diethyl ether, benzene, and chloroform.[13]
The chemical stability of Thiotepa is a critical factor influencing its formulation and clinical administration. It is notably unstable in acidic conditions and is sensitive to light. At temperatures exceeding the recommended storage range of 2–8°C, Thiotepa undergoes polymerization, which renders it inactive. While aqueous solutions of 10 mg/mL are stable for up to five days when refrigerated, the compound is more stable in alkaline solutions.[13] This inherent acid instability explains its unsuitability for oral administration and the necessity for parenteral or local instillation routes.[5]
Table 1 provides a consolidated summary of the key identifiers and properties associated with Thiotepa.
Table 1: Thiotepa Drug Identification Summary
Category | Identifier | Value / Description | Source(s) |
---|---|---|---|
Database & Regulatory IDs | DrugBank ID | DB04572 | 1 |
CAS Number | 52-24-4 | 14 | |
ATC Code | L01AC01 | 6 | |
VA Class Code | AN100 (Antineoplastics, Alkylating Agents) | 16 | |
Chemical Data | Type | Small Molecule | 1 |
Chemical Formula | C6H12N3PS | 6 | |
Molecular Weight | 189.22 g/mol | 15 | |
IUPAC Name | N,N',N''-triethylenethiophosphoramide; 1,1',1''-phosphinothioylidynetrisaziridine | 4 | |
Names and Synonyms | Generic Name | Thiotepa | 1 |
Synonyms | Thio-TEPA, Thiophosphamide, Triethylenethiophosphoramide, Tris(1-aziridinyl)phosphine sulfide, thiofosfamide | 4 | |
Abbreviations / Code Names | TSPA, TESPA, NSC-6396, SH-105, WR 45312 | 4 | |
US Brand Names | Tepadina, Tepylute, Thioplex | 1 | |
International Brand Names | Tepadina, Tepylute, Thiotepa Riemser, Girostan, Ledertepa, Onco Thiotepa, Tespamin, Tifosyl | 1 |
Thiotepa is classified as a polyfunctional, organophosphorus alkylating agent, sharing chemical and pharmacological properties with nitrogen mustards.[4] Its cytotoxic and immunosuppressive effects are mediated through a well-defined mechanism of action. The core process involves the in vivo formation of a highly reactive and unstable ethylenimine radical.[5] This radical is the active species that directly attacks cellular DNA.[1]
The primary molecular target of this electrophilic attack is the N7 position of guanine nucleobases within the DNA double helix. By reacting with these sites, Thiotepa induces the formation of covalent intra-strand and inter-strand cross-links.[1] This cross-linking makes the DNA strands unable to uncoil and separate, a process essential for cellular replication and transcription. The functional consequence is profound and multifaceted: the irreparable DNA damage interferes with DNA replication, RNA transcription, and subsequent protein synthesis. This disruption of fundamental cellular processes ultimately results in the induction of apoptosis and the inhibition of tumor cell growth.[1]
A key feature of Thiotepa's pharmacodynamic profile is that its cytotoxic actions are not specific to any particular phase of the cell cycle.[5] This non-cell-cycle-specificity contributes to its broad-spectrum activity against a variety of both slow- and fast-growing tumors.
Beyond its direct antineoplastic activity, Thiotepa possesses significant immunosuppressive properties that are fundamental to its use in the transplantation setting.[1] By suppressing the host immune system, it helps to prevent the rejection of transplanted hematopoietic progenitor cells. Furthermore, when administered via intracavitary routes (e.g., intrapleural, intraperitoneal), Thiotepa elicits a localized inflammatory reaction on serous membranes. This reaction leads to a sclerosing effect, which aids in the control of malignant effusions by promoting the adhesion of the serosal surfaces.[5]
The pharmacokinetic profile of Thiotepa and its primary active metabolite, TEPA, dictates its route of administration, clinical applications, and unique toxicity profile.
Absorption: Thiotepa is not administered orally. Its chemical structure is unstable in the acidic environment of the gastrointestinal tract, leading to unreliable and incomplete absorption. Consequently, all clinical formulations are designed for parenteral (intravenous) or local (intracavitary, intravesical) administration.[5]
Distribution: Following intravenous administration, Thiotepa exhibits a rapid distribution phase, with peak plasma concentrations occurring immediately.[5] It is characterized as a highly lipophilic and lipid-soluble compound, fitting a two-compartment pharmacokinetic model. This high lipophilicity is a critical property, as it enables Thiotepa to readily cross the blood-brain barrier (BBB).[5] This ability to penetrate the CNS is the pharmacokinetic basis for its efficacy in treating primary and metastatic CNS malignancies, such as primary CNS lymphoma and high-grade gliomas.[19] Its main active metabolite, TEPA, also crosses the BBB, and notably, concentrations of TEPA in the cerebrospinal fluid (CSF) have been shown to exceed those of the parent drug.[5] The volume of distribution (
Vd) is extensive, ranging from 41 to 75 L/m², and plasma protein binding is low, reported to be between 8% and 29%.[5]
Metabolism: Thiotepa undergoes rapid and extensive hepatic metabolism. The principal metabolic pathway is oxidative desulfuration, which is mediated primarily by the Cytochrome P450 enzymes CYP3A4 and CYP2B6.[5] This process generates the major active metabolite,
N,N',N''-triethylenephosphoramide (TEPA). TEPA itself is a potent alkylating agent and contributes significantly to the overall therapeutic and toxic effects of the drug. A secondary metabolic pathway involves conjugation with glutathione to form the thiotepa-mercapturate metabolite.[5] The central role of CYP3A4 and CYP2B6 in its metabolism is the basis for several clinically significant drug-drug interactions.
Excretion: Excretion of Thiotepa and its metabolites occurs through multiple routes. Renal excretion of the unchanged parent drug is minimal. In urine, only about 0.5% of a dose is excreted as thiotepa and its minor metabolite monochlorotepa, while about 11% is excreted as TEPA and thiotepa-mercapturate.[5] One study using a radiolabeled dose reported a total urinary excretion of 63% of the administered radioactivity, indicating that metabolites account for the vast majority of renal clearance.[1]
A unique and clinically important route of excretion, especially with high-dose therapy, is through the skin via sweat.[5] Although the exact percentage of the total dose eliminated via this pathway is unknown, it is sufficient to cause the characteristic cutaneous toxicities associated with the drug.[20] No information is available regarding fecal excretion.[5]
The pharmacokinetic distinction between Thiotepa and its active metabolite TEPA is critical for clinical practice. The longer half-life of TEPA means that its biological effects, both therapeutic and toxic, persist long after the parent drug has been cleared from circulation. This necessitates prolonged monitoring for adverse effects following administration. Table 2 provides a comparative summary of the key pharmacokinetic parameters.
Table 2: Key Pharmacokinetic Parameters of Thiotepa and its Active Metabolite (TEPA)
Parameter | Thiotepa | TEPA (Active Metabolite) | Source(s) |
---|---|---|---|
Oral Bioavailability | Unreliable / Not used orally | N/A | 5 |
Plasma Protein Binding | 8–29% | Data not available | 5 |
Volume of Distribution (Vd) | 41–75 L/m² | Data not available | 5 |
Blood-Brain Barrier | Crosses | Crosses; CSF concentrations can exceed parent drug | 5 |
Primary Metabolism | Hepatic (CYP3A4, CYP2B6 oxidative desulfuration; Glutathione conjugation) | N/A | 5 |
Terminal Half-life (t1/2) | 1.4–3.7 hours | 4.9–17.6 hours | 5 |
Clearance | 11.4–23.2 L/h/m² | Data not available | 5 |
Primary Excretion Route | Metabolism; minor renal excretion; significant excretion via skin/sweat | Renal (as metabolite) | 5 |
Thiotepa has a remarkably long regulatory history that reflects the evolution of cancer therapy over more than half a century. It was first approved by the U.S. FDA in 1959 as a treatment for several types of solid cancers, marking it as one of the earliest cytotoxic agents to enter the clinical armamentarium.[10]
Over the subsequent decades, as manufacturing practices and clinical needs evolved, new formulations received approval. Thioplex, a lyophilized powder for injection, was approved in 1994.[10] This was followed by the approval of
Tepadina, another lyophilized powder formulation, in 2017.[10] The Tepadina approval was significant as it formally added a new indication to the label: its use as part of a conditioning regimen to reduce the risk of graft rejection in pediatric patients with class 3 beta-thalassemia undergoing allogeneic HSCT.[11] This marked a pivotal shift in the drug's recognized role towards specialized transplantation medicine.
This shift was further solidified by its designation as an Orphan Drug by both the European Medicines Agency (EMA) in January 2007 and the FDA in April 2007 for its use as a conditioning treatment prior to HSCT.[2] This designation acknowledged its critical importance in treating rare diseases and conditions requiring transplantation, providing regulatory and commercial incentives for its continued development in this niche area.
Most recently, the focus of innovation has been on improving the drug's handling and administration. In June 2024, the FDA approved Tepylute, a novel ready-to-dilute liquid formulation, for the treatment of breast and ovarian cancer.[3] This was followed by the approval of a multi-dose vial of Tepylute in early 2025, further enhancing its practicality in a clinical setting.[12] This regulatory trajectory—from a broad-spectrum palliative agent to a specialized orphan drug with modernized formulations—demonstrates a successful lifecycle management strategy for a legacy compound, where innovation in drug delivery and indication expansion has sustained its clinical relevance.
The evolution of Thiotepa's pharmaceutical formulations has been driven by the need to improve safety, accuracy, and efficiency in the clinical pharmacy setting.
Lyophilized Powders (e.g., Thioplex, Tepadina): For decades, Thiotepa was available only as a lyophilized (freeze-dried) powder in vials, typically containing 15 mg or 100 mg of the drug.[8] These formulations require a multi-step preparation process that begins with reconstitution using a sterile diluent, such as sterile water for injection.[23] This process has been described as "complicated and time-consuming" and presents several challenges.[10] Manual reconstitution increases the risk of occupational exposure to a potent cytotoxic agent through aerosolization or spills. It also introduces potential for calculation and measurement errors, which can lead to incorrect dosing. Furthermore, the stability of the reconstituted solution is limited; for example, reconstituted Tepadina is stable for only 8 hours under refrigeration.[27]
Ready-to-Dilute Liquid (Tepylute): The approval of Tepylute represents a significant practical advancement. This formulation is supplied as a stable liquid solution, typically at a concentration of 10 mg/mL, that is ready for immediate dilution in an appropriate infusion bag.[10] By eliminating the manual reconstitution step, Tepylute directly addresses the primary challenges of the older formulations. It is intended to provide "easier preparation and dosing," reduce manual compounding time, and improve dosing accuracy.[10] The approval of a multi-dose vial that is stable for 14 days when properly stored provides unprecedented scheduling flexibility for healthcare providers and can significantly reduce drug wastage.[24] This shift in formulation is not merely a matter of convenience; it is a tangible improvement in the quality and safety of medication delivery, benefiting both pharmacy staff and patients by ensuring more precise and safer administration of a high-risk drug. Key manufacturers and distributors in this evolving market include Shorla Oncology, the developer of Tepylute; Adienne Pharma & Biotech, the NDA-holder for Tepadina; and Amneal Biosciences, a major distributor and marketer of generic and branded Thiotepa formulations.[10]
The clinical utility of Thiotepa has evolved significantly since its introduction. While it retains its original FDA-approved indications for certain solid tumors, its most vital role in contemporary medicine is as a component of high-dose conditioning regimens for HSCT.
These indications represent the "traditional" uses of Thiotepa, primarily in a palliative setting. While often superseded by newer therapies, they remain part of its approved labeling.[29]
Thiotepa is indicated for the palliative treatment of adenocarcinoma of the breast and ovary.[1] The newest formulation, Tepylute, is specifically approved for this use.[10] Clinical evidence supports its activity, particularly in challenging scenarios. A retrospective study evaluating intravenous (IV) Thiotepa (40 mg/m²) for leptomeningeal carcinomatosis secondary to metastatic breast cancer (MBC) found the regimen to be well-tolerated and effective, with a partial response observed in 4 of 13 patients and a 6-month survival rate of 69%.[31] Its ability to cross the BBB makes it a logical choice for CNS metastases.[19] Another study of a vinorelbine-thiotepa combination in heavily pretreated MBC patients demonstrated a median progression-free survival (PFS) of 4.4 months (3.6 months in those with CNS metastases), indicating meaningful activity.[19] Ongoing research continues to explore its utility, with trials investigating intrathecal Thiotepa in combination with methotrexate for MBC with leptomeningeal spread.[32]
For ovarian cancer, early-phase trials have explored high-dose Thiotepa in consolidation regimens with stem cell rescue for patients in complete remission.[34] One study of a high-dose combination of busulfan, melphalan, and Thiotepa (BUMELTT) followed by autologous stem cell rescue demonstrated an 18-month overall survival of 87% for patients treated while in complete remission and showed particular benefit in platinum-sensitive disease, with outcomes equivalent to other high-dose regimens.[35]
Thiotepa was a pioneering agent in urologic oncology, opening the era of intravesical chemotherapy for superficial bladder cancer.[36] It is administered directly into the bladder to exert a local cytotoxic effect.[7] Early studies demonstrated that intravesical Thiotepa could completely destroy existing low-stage tumors in approximately one-third of patients.[37]
However, its role in preventing long-term tumor recurrence has been questioned.[37] Multiple large reviews and controlled trials have established that while Thiotepa reduces short-term recurrence by an average of 12-14%, it is significantly less effective than intravesical immunotherapy with Bacillus Calmette-Guérin (BCG).[38] BCG provides superior long-term recurrence prevention and, unlike Thiotepa, has been shown to reduce the risk of disease progression to muscle-invasive cancer.[37] When compared to another intravesical chemotherapeutic, Mitomycin C, Thiotepa has shown generally equal efficacy, although some studies suggest a modest superiority for Mitomycin C.[36] Today, Mitomycin C is often preferred over Thiotepa for this indication due to its larger molecular size, which results in less systemic absorption from the bladder and a correspondingly lower risk of significant myelosuppression.[36]
Thiotepa is indicated for controlling intracavitary effusions (pleural, pericardial, and peritoneal) that are secondary to diffuse or localized neoplastic diseases.[5] When instilled into a body cavity, it exerts not only a direct cytotoxic effect on tumor cells but also an inflammatory, sclerosing effect on the serous membranes, which helps to obliterate the space and prevent fluid re-accumulation.[5]
The most important and dynamic application of Thiotepa in modern oncology is as a component of high-dose conditioning regimens prior to both autologous and allogeneic HSCT. Its combination of potent myelosuppressive and immunosuppressive activity, along with its excellent CNS penetration, makes it an ideal agent for regimens designed to eradicate residual malignant cells (including those in sanctuary sites like the CNS) and ablate the host immune system to prevent graft rejection.[1]
The clinical evidence strongly supports the role of Thiotepa-based conditioning, often demonstrating superiority or non-inferiority to traditional standards of care.
Research into Thiotepa remains active, with a focus on optimizing its use in conditioning regimens and expanding its applications. Numerous clinical trials are currently recruiting or active. Key areas of investigation include its use in combination with targeted agents like venetoclax for high-risk AML and myelodysplastic syndrome (MDS), refining its role in preventing CNS relapse in high-risk diffuse large B-cell lymphoma, and its application in conditioning for non-malignant disorders.[43] Many of these trials explore novel combinations with other chemotherapy drugs such as fludarabine, melphalan, carboplatin, and cytarabine, seeking to enhance efficacy while managing toxicity.[46]
Thiotepa's potent therapeutic effects are accompanied by a significant and well-defined toxicity profile. Its therapeutic index is narrow, and successful clinical use depends on a thorough understanding of its risks and the implementation of proactive management strategies.
The FDA label for Thiotepa includes two critical boxed warnings that underscore its most severe risks.[11]
Severe Myelosuppression: This is the primary and universal dose-limiting toxicity of Thiotepa. The drug is highly toxic to the hematopoietic system, causing profound suppression of all blood cell lineages.[29] At the high doses used in HSCT conditioning, it causes complete and irreversible marrow ablation. Therefore, hematopoietic progenitor (stem) cell transplantation is an absolute requirement to rescue patients from the otherwise fatal complications of prolonged pancytopenia, such as overwhelming infection or catastrophic bleeding.[11] Even at lower doses used for palliative therapy, it can cause severe leukopenia, thrombocytopenia, and anemia.[10] A rapidly falling white blood cell count (e.g., below 3,000/µL) or platelet count (e.g., below 150,000/µL) necessitates immediate dose reduction or discontinuation of therapy.[29] Rigorous and frequent monitoring of hematologic laboratory parameters is mandatory for all patients receiving Thiotepa.[11]
Carcinogenicity: Thiotepa is considered a potential human carcinogen.[11] As a DNA alkylating agent, it is mutagenic and has been demonstrated to be carcinogenic in laboratory animals.[12] In humans, there is an increased risk of developing secondary malignancies following treatment. Cases of myelodysplastic syndromes (MDS) and acute non-lymphocytic leukemia (ANLL) have been reported in patients treated with Thiotepa.[12] Patients must be informed of this long-term risk.
Contraindications:
Warnings and Precautions:
Beyond the boxed warnings, the label includes several other critical warnings. Clinically significant hypersensitivity reactions, including anaphylaxis, can occur and require immediate discontinuation of the drug and appropriate medical intervention.24 Other major warnings that require careful monitoring and management include cutaneous toxicity, hepatic veno-occlusive disease (VOD), central nervous system (CNS) toxicity, and embryo-fetal toxicity.53
Thiotepa can cause a wide range of adverse reactions affecting nearly every organ system. The most common toxicities are a direct consequence of its effect on rapidly dividing cells. Table 3 provides a consolidated overview of adverse reactions compiled from clinical trial data and postmarketing experience.
Table 3: Comprehensive List of Adverse Reactions to Thiotepa by Frequency and System Organ Class
System Organ Class | Very Common (≥10%) | Common (1-10%) | Serious / Clinically Significant (Frequency Varies) |
---|---|---|---|
Blood and Lymphatic | Neutropenia, Anemia, Thrombocytopenia, Febrile Neutropenia | - | Bone marrow ablation, Febrile bone marrow aplasia, Pancytopenia |
Infections | Cytomegalovirus (CMV) infection, Sepsis, General infections | - | Life-threatening infections due to immunosuppression |
Gastrointestinal | Mucositis (Stomatitis, Esophagitis), Nausea, Vomiting, Diarrhea, Abdominal pain | Constipation, Dyspepsia | GI perforation, Ileus, Colitis, GI hemorrhage |
Hepatobiliary | Elevated ALT, Elevated AST, Elevated Bilirubin | - | Hepatic Veno-Occlusive Disease (VOD/SOS), Jaundice |
Skin and Subcutaneous | Rash, Pruritus, Alopecia | Erythema, Blistering, Desquamation, Hyperpigmentation | Severe cutaneous reactions (e.g., Stevens-Johnson Syndrome), Contact dermatitis |
Nervous System | Dizziness, Headache | Paresthesia, Cognitive disorder, Confusion, Encephalopathy | Fatal encephalopathy, Seizures, Coma, Cerebral hemorrhage, Leukoencephalopathy |
Renal and Urinary | Hematuria | Dysuria, Urinary retention | Chemical cystitis, Hemorrhagic cystitis (intravesical use) |
General Disorders | Pyrexia (Fever), Asthenia (Weakness), Chills, Pain | Fatigue | Multi-organ failure, Anaphylaxis, Hypersensitivity reactions |
Cardiac | - | - | Cardiorespiratory arrest, Congestive heart failure, Bradycardia, Pericardial effusion |
Metabolism | Anorexia | - | Electrolyte abnormalities (Hypokalemia, Hypocalcemia) |
Psychiatric | Mental status changes | Anxiety, Delirium, Hallucinations, Agitation | - |
Neoplasms | - | - | Secondary malignancies (MDS, ANLL) |
Reproductive | - | Amenorrhea, Impaired spermatogenesis | Infertility |
Sources: [10]
A distinctive toxicity of high-dose Thiotepa is a severe cutaneous reaction. The pathophysiology is directly linked to its pharmacokinetic profile: Thiotepa and its active metabolites are partially excreted through the skin in sweat.[5] This leads to high local concentrations of the cytotoxic agent on the skin surface, causing a direct toxic effect.
The clinical presentation is a consistent pattern of diffuse erythema (redness), which can progress to blistering, exfoliation (peeling), and desquamation (shedding of skin).[20] It is often accompanied by significant pruritus (itching) and can result in a bronze-like or tan hyperpigmentation that fades over several weeks to months. The toxicity is characteristically more severe in intertriginous and occluded areas where sweat accumulates, such as the groin, axillae, neck, and under adhesive dressings or electrocardiogram pads.[12]
Management is primarily prophylactic and relies on patient education. The following measures are recommended during Thiotepa administration and for at least 48 hours after the final dose:
Hepatic VOD, also known as Sinusoidal Obstruction Syndrome (SOS), is a serious and potentially fatal complication that can occur in patients receiving high-dose Thiotepa, particularly when it is used in combination with other hepatotoxic alkylating agents like busulfan and cyclophosphamide in the HSCT setting.[22] It involves damage to the sinusoidal endothelial cells in the liver, leading to obstruction and subsequent liver failure. Close monitoring for signs of VOD—such as rapid weight gain, painful hepatomegaly, ascites, and rising bilirubin levels—is essential.[24]
Given its ability to cross the BBB, Thiotepa can cause significant CNS toxicity, which appears to be dose-dependent. In patients treated with high doses, fatal encephalopathy has been reported.[12] A spectrum of other CNS toxicities can occur, including headache, apathy, confusion, disorientation, amnesia, hallucinations, somnolence, seizures, and coma.[12] Any new neurologic symptom should be investigated immediately.
Thiotepa is a potent teratogen. It can cause fetal harm when administered to a pregnant woman and has demonstrated teratogenicity in animal studies at doses well below the human therapeutic dose on a body-surface area basis.[30] Therefore, pregnancy must be avoided. Females of reproductive potential must be advised to use highly effective contraception during treatment and for at least 6 months after the final dose. Males with female partners of reproductive potential must use effective contraception during treatment and for at least 1 year after the final dose, due to the drug's effects on spermatogenesis.[57] Thiotepa can cause amenorrhea in females and interfere with sperm production in males, potentially leading to permanent infertility.[29]
Navigating potential drug-drug interactions is crucial for the safe and effective use of Thiotepa. Interactions can be pharmacokinetic, affecting drug exposure, or pharmacodynamic, leading to additive toxicity or antagonistic effects.
Thiotepa is extensively metabolized by the hepatic enzymes CYP3A4 and CYP2B6.[5] Concomitant use of drugs that modulate these enzymes can significantly alter the plasma concentrations of Thiotepa and its active metabolite, TEPA.
Pharmacodynamic interactions occur when two drugs have additive or opposing effects on the body.
Table 4 summarizes the most clinically important drug interactions and provides management recommendations.
Table 4: Key Drug Interactions with Thiotepa and Clinical Recommendations
Interacting Agent / Class | Mechanism of Interaction | Potential Effect | Clinical Recommendation | Source(s) |
---|---|---|---|---|
Strong CYP3A4 Inhibitors (e.g., itraconazole, ritonavir) | Pharmacokinetic (Decreased Thiotepa metabolism) | Increased Thiotepa plasma concentration and risk of severe toxicity | Avoid co-administration. Consider alternative medications. | 1 |
Strong CYP3A4 Inducers (e.g., rifampin, phenytoin) | Pharmacokinetic (Increased Thiotepa metabolism) | Decreased Thiotepa plasma concentration and potential loss of efficacy | Avoid co-administration. Consider alternative medications. | 1 |
Other Myelosuppressive Agents (e.g., other chemotherapy, radiation) | Pharmacodynamic (Additive toxicity) | Profound and prolonged myelosuppression | Avoid simultaneous use. Monitor blood counts closely. | 54 |
Anticoagulants / Antiplatelets (e.g., acenocoumarol, abciximab) | Pharmacodynamic (Additive effect) | Increased risk of serious bleeding | Use with extreme caution. Monitor closely for signs of bleeding. | 1 |
Live or Attenuated Vaccines (e.g., MMR, yellow fever) | Pharmacodynamic (Immunosuppression) | Diminished vaccine response and risk of disseminated infection | Contraindicated during and for a period after Thiotepa therapy. | 48 |
Succinylcholine | Pharmacokinetic (Inhibition of pseudocholinesterase) | Prolonged apnea and neuromuscular blockade | Use with caution. Inform anesthesiologist of Thiotepa use. | 29 |
The dosing of Thiotepa is highly individualized based on the indication, route of administration, patient's body weight or body surface area, and hematologic status.
The following are general dosing guidelines derived from FDA-approved labeling. Doses must be carefully adjusted based on patient tolerance and response, particularly blood counts.[11]
Thiotepa is a potent cytotoxic drug that requires careful handling to protect healthcare personnel from exposure.
Thiotepa stands as a testament to the enduring utility of foundational chemotherapeutic agents in an era increasingly dominated by targeted therapies and immunotherapies. Its clinical journey over more than 60 years illustrates a remarkable evolution, transitioning from a broad-spectrum palliative agent to a highly specialized, indispensable tool in transplantation oncology. This report has synthesized a vast body of evidence to construct a comprehensive profile of the drug, revealing several key themes.
First, Thiotepa's clinical identity is now twofold. While it retains its historical, FDA-approved indications for breast, ovarian, and bladder cancers, its clinical impact in these areas has been largely superseded by more effective and less toxic alternatives. Its true modern value lies in its role as a cornerstone of high-dose conditioning regimens for HSCT. This "rebirth" is a direct result of its unique pharmacological profile: the combination of potent, non-cell-cycle-specific myelosuppression and excellent CNS penetration makes it uniquely suited to eradicate malignant cells in sanctuary sites, a critical goal in potentially curative transplant procedures. The robust clinical data supporting its superiority over regimens like BEAM in PCNSL and its non-inferiority to TBI in ALL underscore its critical place in the current treatment paradigm.
Second, the therapeutic index of Thiotepa is narrow, and its use is fundamentally defined by a profound but manageable toxicity profile. The direct line from its pharmacokinetics and mechanism of action to its signature adverse effects—myelosuppression from its effect on bone marrow, cutaneous toxicity from its excretion in sweat, and CNS toxicity from its ability to cross the blood-brain barrier—provides a clear, mechanistic framework for understanding its risks. The successful application of Thiotepa is therefore less about novel discovery and more about meticulous clinical management: careful patient selection, aggressive supportive care, proactive toxicity mitigation (e.g., mandatory skin hygiene), and vigilant monitoring for drug interactions and long-term sequelae like secondary malignancies.
Third, the recent regulatory approval of new, ready-to-dilute liquid formulations like Tepylute marks a significant step forward. This innovation in drug delivery, while seemingly simple, addresses long-standing challenges related to the handling of a hazardous cytotoxic agent. By improving the safety, precision, and efficiency of its preparation and administration, these new formulations enhance the overall quality of care and represent a meaningful advancement in the practical application of a legacy drug.
Looking to the future, research on Thiotepa is likely to focus on further optimization rather than new discovery. Key questions remain: Can Thiotepa-based conditioning regimens be refined using pharmacokinetic-guided dosing to minimize inter-patient variability and further improve the balance between efficacy and non-relapse mortality? What are the optimal combination partners for Thiotepa in different disease settings, particularly with the advent of novel targeted and cellular therapies? And critically, how can the long-term risk of secondary malignancies be better predicted, monitored, and mitigated in the growing population of long-term survivors who have received Thiotepa-based conditioning?
In conclusion, Thiotepa is a powerful and clinically relevant therapeutic agent. Its journey from a broadly used palliative drug to a specialized cornerstone of HSCT highlights the dynamic nature of clinical oncology. Its continued study and the ongoing improvements in its formulation and administration ensure that despite its age, Thiotepa will remain a vital component of the oncologic armamentarium for the foreseeable future.
Published at: July 23, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.