A Comprehensive Monograph on Temozolomide: Pharmacology, Clinical Efficacy, and Mechanisms of Resistance
Executive Summary
Temozolomide is an oral and intravenous alkylating agent that has fundamentally altered the therapeutic landscape for patients with high-grade malignant gliomas. As an imidazotetrazine derivative, it functions as a prodrug, undergoing non-enzymatic conversion at physiological pH to a reactive species that methylates tumor cell DNA, inducing cytotoxic lesions that lead to apoptosis. Its excellent oral bioavailability and ability to penetrate the blood-brain barrier make it uniquely suited for treating central nervous system malignancies.
The clinical utility of Temozolomide was definitively established by the landmark 2005 phase III trial by Stupp et al., which demonstrated that adding concomitant and adjuvant Temozolomide to standard radiotherapy significantly improved median and long-term survival for patients with newly diagnosed glioblastoma (GBM). This regimen, now known as the "Stupp protocol," was rapidly adopted as the global standard of care. The drug has also proven its efficacy in anaplastic astrocytoma (AA), first in the refractory/recurrent setting and more recently as an adjuvant therapy for newly diagnosed, IDH-mutant tumors.
Despite its success, the efficacy of Temozolomide is profoundly limited by chemoresistance. The primary mechanism of resistance is the expression of the DNA repair enzyme O⁶-methylguanine-DNA methyltransferase (MGMT), which directly reverses the drug's most cytotoxic DNA lesion. The epigenetic silencing of the MGMT gene via promoter methylation prevents the production of this enzyme, rendering tumors sensitive to Temozolomide. Consequently, MGMT promoter methylation status has emerged as the most critical predictive biomarker in neuro-oncology, stratifying patients into responder and non-responder populations. However, resistance is a complex phenomenon, with MGMT-independent pathways, including deficiencies in the Mismatch Repair (MMR) system and hyperactivity of the Base Excision Repair (BER) pathway, also playing significant roles.
The safety profile of Temozolomide is characterized primarily by a predictable and manageable myelosuppression, which is the principal dose-limiting toxicity. Other significant adverse events include nausea, fatigue, hepatotoxicity, and a long-term risk of secondary malignancies such as myelodysplastic syndrome. Current and future research is intensely focused on strategies to overcome the multifaceted mechanisms of resistance, employing novel drug combinations, advanced delivery systems, and biomarker-driven therapeutic approaches to improve outcomes for patients with these devastating brain cancers.
Identification and Physicochemical Properties
This section provides a definitive catalog of the nomenclature, identifiers, and core chemical and physical characteristics of the Temozolomide molecule, establishing a foundational reference for its clinical and research applications.
Nomenclature and Identifiers
Temozolomide is recognized by a variety of names and unique identifiers across chemical, pharmaceutical, and regulatory databases, reflecting its journey from a research compound to a globally utilized chemotherapy agent.
- Generic Name: Temozolomide [1]
- Brand Names: The drug is marketed globally under several brand names, most prominently as Temodar® in the United States and Temodal® in the European Union and other international markets.[3] Other registered trade names include Temomedac and Temcad.[1]
- Systematic (IUPAC) Name: The formal chemical name according to IUPAC nomenclature is 4-methyl-5-oxo-2,3,4,6,8-pentazabicyclo[4.3.0]nona-2,7,9-triene-9-carboxamide.[6] An alternative and widely used systematic name is 3,4-dihydro-3-methyl-4-oxoimidazo[5,1-d]-1,2,3,5-tetrazine-8-carboxamide.[7]
- Synonyms: During its development and in various research contexts, Temozolomide has been referred to by numerous synonyms and code names. These include Methazolastone, CCRG 81045, M&B 39831, and NSC 362856.[7]
- Key Identifiers:
- CAS Number: 85622-93-1 [9]
- DrugBank ID: DB00853 [1]
- PubChem Compound ID (CID): 5394 [6]
- ChEMBL ID: CHEMBL810 [6]
- UNII (Unique Ingredient Identifier): YF1K15M17Y [6]
Chemical Structure and Formula
Temozolomide is a small molecule belonging to the imidazotetrazine class of compounds and is classified as a second-generation alkylating agent.[2]
- Molecular Formula: C6H6N6O2 [10]
- Molecular Weight: 194.15 g/mol [9]
Physical and Chemical Properties
The physical and chemical characteristics of Temozolomide dictate its formulation, stability, handling requirements, and biological behavior.
- Appearance: The pure substance is a white to light tan or light pink powder.[7]
- Solubility: It is slightly soluble in water (approximately 3.5 g/L) and aqueous acids.[6] For laboratory use, it is soluble in dimethyl sulfoxide (DMSO) at concentrations up to 25 mg/mL and in dimethylformamide (DMF) at 5 mg/mL.[7]
- Stability and Storage: The stability of Temozolomide is dependent on its formulation and environment.
- Oral Capsules: Should be stored at controlled room temperature, between 20°C to 25°C (68°F to 77°F).[17]
- Bulk Chemical: For long-term stability (at least 2 years), the powder should be stored at 2-8°C, protected from light and moisture.[7]
- Reconstituted IV Solution: The intravenous solution, once reconstituted, is stable for 14 hours at room temperature (25°C), which includes the infusion time.[19]
- Melting Point: The compound decomposes upon heating at 212°C (414°F).[6]
- Hazard Information: Temozolomide is classified as a hazardous substance with significant biological risks. According to the Globally Harmonized System (GHS), it is designated with the hazard statements H350 (May cause cancer), H360 (May damage fertility or the unborn child), and H302 (Harmful if swallowed).[7] Furthermore, it has been discovered to be an explosive material, tentatively assigned as UN Class 1.[6]
The inherent chemical instability and toxicity of the Temozolomide molecule have direct and critical implications for its clinical use and handling. The GHS classifications for carcinogenicity and reproductive toxicity, combined with its explosive nature, necessitate stringent safety protocols. This is reflected in the FDA-approved labeling and administration guidelines, which explicitly warn both healthcare professionals and patients that the oral capsules must never be opened, chewed, or dissolved but must be swallowed whole.[19] This precaution serves to prevent aerosolization of the hazardous powder and direct contact with skin or mucous membranes, thereby protecting both the patient and caregivers from unintended exposure. This provides a clear example of how the fundamental chemical properties of a drug directly inform and mandate crucial clinical safety practices.
Formulations and Strengths
Temozolomide is available in both oral and intravenous formulations to accommodate different clinical scenarios.
- Oral Capsules: The drug is supplied in hard capsules of various strengths: 5 mg, 20 mg, 100 mg, 140 mg, 180 mg, and 250 mg.[17] While some brand-name Temodar® capsule strengths have been discontinued, generic versions remain available.[17]
- Intravenous (IV) Injection: For patients who cannot take oral medication, Temozolomide is available as a lyophilized powder for reconstitution in single-dose glass vials, each containing 100 mg of the active drug.[23]
Table 1: Drug Identification and Chemical Properties
Property | Value |
---|
Generic Name | Temozolomide |
Primary Brand Names | Temodar, Temodal |
Drug Class | Alkylating Agent, Imidazotetrazine derivative |
CAS Number | 85622-93-1 |
DrugBank ID | DB00853 |
Molecular Formula | C6H6N6O2 |
Molecular Weight | 194.15 g/mol |
Appearance | White to light pink powder |
Key Solvents | DMSO, DMF, Water (slightly) |
Storage Conditions | Capsules: Room Temperature; Chemical: 2-8°C, protect from light/moisture |
GHS Hazard Codes | H350 (Carcinogenic), H360 (Reproductive Toxin), H302 (Harmful) |
Clinical Pharmacology
The clinical pharmacology of Temozolomide describes its interaction with the human body, encompassing its mechanism of action (pharmacodynamics) and its absorption, distribution, metabolism, and excretion (pharmacokinetics). These properties collectively explain its therapeutic efficacy and define its clinical application.
A. Mechanism of Action (Pharmacodynamics)
Temozolomide exerts its anticancer effects through a unique, multi-step process involving activation, DNA alkylation, and the induction of programmed cell death.
Prodrug Activation
Temozolomide is a prodrug, a biologically inert compound that is converted into its active form within the body.[1] A key pharmacological advantage of Temozolomide is that this activation occurs through a spontaneous, non-enzymatic chemical reaction, rather than requiring metabolic processing by the liver.[10] At physiological pH (neutral to slightly alkaline, pH > 7), the imidazotetrazine ring of the Temozolomide molecule opens via hydrolysis to form the active metabolite, 3-methyl-(triazen-1-yl)imidazole-4-carboxamide, commonly known as MTIC.[1] This independence from the cytochrome P450 enzyme system minimizes the potential for many common drug-drug interactions, leading to a more predictable pharmacokinetic profile.
This pH-dependent activation mechanism may also confer a degree of tumor-selective activity. Malignant tissues, including brain tumors, often exhibit a slightly more alkaline extracellular pH compared to surrounding healthy tissue.[1] Because the hydrolysis of Temozolomide to its active form is favored under these more basic conditions, it is plausible that the conversion to the cytotoxic MTIC is accelerated within the tumor microenvironment itself. This creates a scenario where the drug's activation is inherently biased toward its target site, potentially enhancing its therapeutic index. Following its formation, the unstable MTIC molecule rapidly decomposes to release two components: 5-aminoimidazole-4-carboxamide (AIC), a natural purine precursor, and the highly reactive methyl diazonium cation (
CH3N2+), which is the ultimate DNA-alkylating species.[1]
DNA Alkylation and Cytotoxicity
The therapeutic effect of Temozolomide is driven by the ability of the methyl diazonium cation to covalently transfer a methyl group to the DNA of cancer cells.[1] This process, known as alkylation, creates DNA adducts at several positions on the purine bases, primarily:
- N7 position of guanine (N7-MeG): This is the most frequent modification, accounting for approximately 70% of all methylation events.
- N3 position of adenine (N3-MeA): This site accounts for about 9% of the adducts.
- O6 position of guanine (O6-MeG): This is a less frequent modification, representing only about 6% of the total alkylation events.[1]
Induction of Cell Death
The cytotoxic outcome of these DNA lesions is not uniform; it is critically dependent on the cell's intrinsic DNA repair capabilities. While the N7-MeG and N3-MeA adducts are the most numerous, they are generally considered less cytotoxic because they are efficiently recognized and removed by the ubiquitous Base Excision Repair (BER) pathway.[26]
The therapeutic efficacy of Temozolomide hinges almost entirely on the fate of the far less common O6-MeG lesion.[6] This specific adduct is a "ticking time bomb" for the cell. Its repair is handled exclusively by a specialized DNA repair enzyme called O⁶-methylguanine-DNA methyltransferase (MGMT). If the MGMT enzyme is present and active, it removes the methyl group, repairing the DNA and rendering the cell resistant to the drug's effects.
However, if the tumor cell lacks functional MGMT, the O6-MeG lesion persists through DNA replication. During this process, DNA polymerase incorrectly pairs the O6-methylguanine with a thymine (T) base instead of the correct cytosine (C) base. This O6-MeG:T mismatch is then recognized by the cell's DNA Mismatch Repair (MMR) system.[29] The MMR system attempts to "correct" this error, but because the original O6-MeG lesion remains on the template strand, these attempts are futile. This leads to repeated cycles of excision and repair, which ultimately result in persistent DNA single- and double-strand breaks. These irreparable breaks trigger a cell cycle arrest at the G2/M checkpoint and initiate the cascade of programmed cell death, or apoptosis.[11] Some evidence also suggests that this process can induce autophagy, another form of cellular self-destruction.[7] This complex cascade explains why the presence or absence of a single repair enzyme, MGMT, is the principal determinant of a tumor's sensitivity or resistance to Temozolomide.
Immunomodulatory Effects
Beyond its direct cytotoxic effects, there is growing interest in the immunomodulatory properties of Temozolomide. The drug's known myelosuppressive side effect leads to a reduction in circulating lymphocytes, a state known as lymphodepletion. Counterintuitively, this process may enhance antitumor immunity. By depleting the pool of lymphocytes, Temozolomide may preferentially eliminate immunosuppressive cells, such as regulatory T-cells (Tregs), within the tumor microenvironment. This can shift the balance toward a more effective, tumor-specific immune response, potentially creating a more favorable setting for the activity of concurrent or subsequent immunotherapies.[1]
B. Pharmacokinetics
The pharmacokinetic profile of Temozolomide describes its movement into, through, and out of the body (ADME: Absorption, Distribution, Metabolism, and Excretion).
- Absorption: Following oral administration, Temozolomide is absorbed rapidly and completely from the gastrointestinal tract, exhibiting nearly 100% bioavailability.[1] Under fasting conditions, the time to reach maximum plasma concentration (Tmax) is approximately one hour.[1]
- Effect of Food: The absorption of oral Temozolomide is significantly affected by the presence of food. Administration with a high-fat meal has been shown to reduce the peak plasma concentration (Cmax) by 32% and the total drug exposure (Area Under the Curve, AUC) by 9%. It also delays the time to reach peak concentration by twofold (from 1 hour to 2.25 hours).[1] This pharmacokinetic interaction is the basis for the clinical recommendation that patients take the medication on an empty stomach to ensure maximal and consistent absorption.[19]
- Distribution: Temozolomide distributes widely throughout the body. It has a relatively low apparent volume of distribution of approximately 0.4 L/kg and exhibits low binding to plasma proteins (about 15%).[1] A critical feature for its use in neuro-oncology is its ability to readily cross the blood-brain barrier (BBB). Studies have shown that the concentration of Temozolomide in the cerebrospinal fluid (CSF) is approximately 30% of the concentration measured in the blood plasma, confirming that the drug achieves therapeutically relevant levels at its intended site of action in the brain.[1]
- Metabolism and Elimination: As previously described, the primary "metabolism" of Temozolomide is its non-enzymatic chemical conversion to the active metabolite MTIC. It has a short elimination half-life of approximately 1.8 hours.[6] The parent drug and its metabolites, including AIC, are primarily cleared from the body through renal excretion into the urine.[6]
Table 2: Summary of Pharmacokinetic Parameters
Parameter | Value |
---|
Bioavailability (Oral) | ~100% |
Time to Peak Plasma (Tmax) | ~1 hour (fasting) |
Effect of High-Fat Meal | ↓ Cmax by 32%, ↓ AUC by 9% |
Plasma Protein Binding | ~15% |
CNS Penetration (CSF:Plasma Ratio) | ~30% |
Elimination Half-Life | ~1.8 hours |
Primary Route of Elimination | Renal (Urine) |
Clinical Efficacy in Malignant Gliomas
The role of Temozolomide in modern neuro-oncology is defined by evidence from pivotal clinical trials that have established its efficacy in the most aggressive forms of brain cancer: glioblastoma and anaplastic astrocytoma.
A. Glioblastoma (WHO Grade IV): The Stupp Protocol - A Paradigm Shift in Treatment
The introduction of Temozolomide into the first-line treatment of glioblastoma (GBM) represents one of the most significant advances in neuro-oncology in recent decades.
Background and the Pivotal EORTC/NCIC Trial
Prior to 2005, the standard of care for adults with newly diagnosed GBM consisted of surgical resection to the maximal extent feasible, followed by adjuvant radiotherapy (RT).[33] This approach yielded dismal outcomes, with a median overall survival of approximately 12 months and very few patients surviving beyond two years.[33]
This prognosis was profoundly changed by a landmark phase III randomized clinical trial conducted by the European Organisation for Research and Treatment of Cancer (EORTC) and the National Cancer Institute of Canada (NCIC) Clinical Trials Group, led by Dr. Roger Stupp and published in 2005.[35] The trial (NCT00006353) enrolled 573 patients with newly diagnosed GBM and randomized them to receive either standard radiotherapy alone or radiotherapy combined with both concomitant and adjuvant Temozolomide.[33]
The combination regimen, which became known as the "Stupp protocol," consisted of two phases:
- Concomitant Phase: Patients received focal radiotherapy (a total dose of 60 Gy delivered in 30 fractions over 6 weeks) concurrently with continuous daily oral Temozolomide at a dose of 75 mg/m²/day for the entire duration of radiotherapy (42-49 days).[32]
- Adjuvant Phase: Following a four-week rest period, patients received up to six 28-day cycles of adjuvant Temozolomide, administered at a dose of 150-200 mg/m²/day for the first five days of each cycle.[35]
Key Efficacy Outcomes and Clinical Impact
The results of the trial were practice-changing, demonstrating a statistically significant and clinically meaningful survival benefit for the group receiving Temozolomide.[33] At a median follow-up of 28 months, the key findings were:
- Median Overall Survival (OS): The median OS was extended by 2.5 months, from 12.1 months in the radiotherapy-alone arm to 14.6 months in the radiotherapy-plus-Temozolomide arm. This corresponded to a 37% reduction in the risk of death (Hazard Ratio for death: 0.63; 95% Confidence Interval [CI], 0.52 to 0.75; p<0.001).[33]
- Two-Year Survival Rate: The benefit of Temozolomide was even more pronounced in long-term survival. The rate of patients alive at two years more than doubled, from 10.4% with radiotherapy alone to 26.5% with the combination therapy.[33]
- Five-Year Survival Rate: A subsequent follow-up analysis of the trial confirmed the durability of this benefit. The five-year survival rate was 9.8% in the Temozolomide group, compared to a mere 1.9% in the radiotherapy-alone group, a five-fold increase.[38]
While a 2.5-month improvement in median survival may appear modest in some oncological contexts, its impact in GBM was profound. The true significance of the Stupp protocol lies not just in shifting the median, but in substantially "fattening the tail" of the survival curve. The dramatic improvements in 2- and 5-year survival rates indicated that the addition of Temozolomide did not simply delay the inevitable for the average patient; rather, it created a new, albeit small, cohort of long-term survivors in a disease where this was previously almost non-existent. This fundamentally redefined the therapeutic goals for GBM, introducing the possibility of durable disease control for a subset of patients and establishing the Stupp protocol as the undisputed global standard of care.[36]
Ongoing Questions: Duration of Adjuvant Therapy
The original Stupp protocol specified six cycles of adjuvant Temozolomide. However, whether extending this duration provides additional benefit remains a subject of debate. Several retrospective studies and clinical experiences have suggested that continuing treatment for 12 or more cycles may improve outcomes, particularly for patients with MGMT-unmethylated tumors who have a poorer prognosis.[40] Conversely, a prospective phase II trial from the Spanish neuro-oncology group (GEINO 14-01) found that extending Temozolomide beyond the sixth cycle offered no progression-free survival benefit and was associated with increased toxicity.[40] As such, the optimal duration of adjuvant therapy beyond the standard six cycles is not definitively established and is often decided on a case-by-case basis.
Numerous clinical trials continue to investigate ways to improve upon the Stupp regimen by adding other agents, including anti-angiogenic drugs like bevacizumab, immunotherapies, and other targeted molecules, though none have yet surpassed the original protocol as the foundational standard of care.[42]
B. Anaplastic Astrocytoma (WHO Grade III)
Temozolomide's role in anaplastic astrocytoma (AA) has also been firmly established, following a developmental path from treating relapsed disease to becoming a key component of first-line adjuvant therapy.
Treatment of Refractory and Recurrent Disease
The initial FDA approval for Temozolomide in 1999 was for the treatment of adult patients with refractory anaplastic astrocytoma.[2] This approval was based on the results of a pivotal multicenter, open-label, phase II trial led by Dr. W.K. Alfred Yung, which evaluated single-agent Temozolomide in patients with AA or anaplastic oligoastrocytoma at their first relapse.[45]
In the intent-to-treat population of 162 patients, the study demonstrated significant antitumor activity:
- Progression-Free Survival (PFS) at 6 months: The primary endpoint was met, with a 6-month PFS rate of 46% (95% CI, 38-54%).
- Median PFS: 5.4 months.
- Median Overall Survival (OS): 13.6 months.
- Objective Response Rate (ORR): An independent review of MRI scans showed an ORR of 35%, which included an 8% complete response rate and a 27% partial response rate. An additional 26% of patients achieved stable disease.
The trial concluded that Temozolomide possessed good single-agent activity with an acceptable and manageable safety profile, establishing it as a critical therapeutic option for patients with recurrent AA.[45]
Adjuvant Therapy for Newly Diagnosed Disease
The success of Temozolomide in the recurrent setting and in first-line GBM provided the rationale for its investigation as part of the initial treatment for newly diagnosed AA. This question was addressed by the large, randomized CATNON trial (NCT00626990), which evaluated different schedules of Temozolomide with radiotherapy in patients with newly diagnosed, 1p/19q non-codeleted anaplastic glioma.[22]
Long-term analysis of this trial revealed that the addition of adjuvant Temozolomide (up to 12 cycles) after completion of radiotherapy significantly improved overall survival in the subset of patients whose tumors harbored an IDH mutation.[49] Interestingly, the addition of concomitant Temozolomide during radiotherapy did not confer an additional survival benefit.[49] Based on this high-level evidence, radiotherapy followed by 12 cycles of adjuvant Temozolomide is now considered a standard of care for newly diagnosed, IDH-mutant anaplastic astrocytoma. This led to a formal update of the FDA label in September 2023, granting a new indication for the adjuvant treatment of adults with newly diagnosed anaplastic astrocytoma.[22] This clinical development history illustrates a logical and successful drug development pathway, progressing from proof-of-concept in a salvage setting to a refined, biomarker-driven role in first-line therapy.
C. Other Investigational Uses
The broad-spectrum alkylating activity of Temozolomide has prompted its investigation in a variety of other malignancies. It has shown activity in combination regimens for advanced pancreatic neuroendocrine tumors and has been studied in metastatic melanoma, aggressive pituitary tumors, and relapsed small cell lung cancer.[6] However, for most of these indications, its use remains experimental and is not part of standard-of-care guidelines.[51]
Table 4: Efficacy Outcomes from the Pivotal Glioblastoma Trial (Stupp et al., 2005)
Outcome Metric | Radiotherapy + Temozolomide (n=287) | Radiotherapy Alone (n=286) | Hazard Ratio (95% CI) / p-value |
---|
Median Overall Survival | 14.6 months | 12.1 months | 0.63 (0.52-0.75) / p<0.001 |
2-Year Overall Survival Rate | 26.5% | 10.4% | Not Applicable |
5-Year Overall Survival Rate | 9.8% | 1.9% | Not Applicable |
Median Progression-Free Survival | 6.9 months | 5.0 months | 0.54 (0.45-0.64) / p<0.001 |
Table 5: Efficacy Outcomes from the Phase II Recurrent Anaplastic Astrocytoma Trial (Yung et al., 1999)
Efficacy Endpoint | Result (ITT Population, n=162) |
---|
Progression-Free Survival (PFS) at 6 Months | 46% (95% CI: 38-54%) |
Median Progression-Free Survival | 5.4 months |
Median Overall Survival | 13.6 months |
Overall Response Rate (ORR) | 35% |
Complete Response (CR) Rate | 8% |
Partial Response (PR) Rate | 27% |
Stable Disease (SD) Rate | 26% |
Dosing, Administration, and Formulation
The effective and safe use of Temozolomide requires strict adherence to indication-specific dosing regimens, careful hematologic monitoring, and proper handling procedures. This section provides a detailed clinical guide to its administration.
General Principles
- Dosing Calculation: All Temozolomide doses are calculated based on the patient's body surface area (BSA) and are expressed in milligrams per square meter (mg/m²).[5]
- Route of Administration: The drug can be administered either orally via capsules or as an intravenous (IV) infusion. The oral and IV formulations are considered bioequivalent when the IV form is infused over a period of 90 minutes, and thus the recommended dose is identical for both routes.[25]
- Oral Administration: To mitigate the common side effects of nausea and vomiting, patients are advised to take the oral capsules on an empty stomach (at least one hour before or two hours after a meal) or at bedtime.[19] Prophylactic use of antiemetic medication is often recommended.[31]
- Capsule Handling: Due to the drug's cytotoxic and hazardous nature, the capsules must be swallowed whole with a glass of water. They should never be opened, chewed, or dissolved to prevent exposure to the powder within.[19]
Recommended Dosing for Approved Indications
The dosing schedule for Temozolomide is highly specific to the indication and the phase of treatment.
- Newly Diagnosed Glioblastoma (Stupp Protocol):
- Concomitant Phase: The recommended dose is 75 mg/m² administered once daily for 42 to 49 consecutive days, concurrently with the full course of focal radiotherapy.[19]
- Maintenance Phase: This phase begins four weeks after the completion of chemoradiotherapy and consists of six 28-day cycles.
- Cycle 1: The dose is 150 mg/m² once daily for Days 1 through 5.
- Cycles 2-6: If Cycle 1 was well-tolerated (based on hematologic and non-hematologic toxicity), the dose may be escalated to 200 mg/m² once daily for Days 1 through 5. If the dose is not escalated at Cycle 2, it should not be increased in subsequent cycles.[19]
- Adjuvant Treatment of Newly Diagnosed Anaplastic Astrocytoma:
- Treatment begins approximately four weeks after the completion of radiotherapy and continues for up to 12 cycles.
- Cycle 1: The dose is 150 mg/m² once daily for Days 1 through 5 of a 28-day cycle.
- Cycles 2-12: The dose is increased to 200 mg/m² once daily for Days 1 through 5, provided there was no significant toxicity in Cycle 1.[5]
- Refractory Anaplastic Astrocytoma:
- Initial Dose (Cycle 1): The starting dose is 150 mg/m² once daily for Days 1 through 5 of a 28-day cycle.
- Subsequent Cycles: The dose may be increased to 200 mg/m² in subsequent cycles if the patient's blood counts at the nadir and at the start of the next cycle meet specified safety criteria.[5]
Hematologic Monitoring and Dose Modifications
Myelosuppression is the primary dose-limiting toxicity of Temozolomide, necessitating a carefully designed administration and monitoring schedule to allow for bone marrow recovery between cycles. The entire 28-day cycle structure, with 5 days of treatment followed by a 23-day rest period, is built around this principle. The nadir, or lowest point, of neutrophil and platelet counts typically occurs late in the cycle, between Day 21 and Day 28.[55] This pharmacokinetic reality is why blood count monitoring at specific time points is a mandatory safety measure.
- Prerequisite for Dosing: Before initiating treatment or starting any new cycle, the patient's Absolute Neutrophil Count (ANC) must be ≥1.5 x 10⁹/L and the platelet count must be ≥100 x 10⁹/L.[1]
- Monitoring Schedule:
- Concomitant Phase (GBM): A Complete Blood Count (CBC) must be obtained weekly.[19]
- Maintenance/Adjuvant Cycles: A CBC must be obtained on Day 22 (or within 48 hours of that day) of each cycle. If counts are low, weekly monitoring is required until they recover to safe levels.[23]
- Dose Interruption and Reduction: The prescribing information provides detailed tables for dose modifications based on the severity of hematologic toxicity. As a general rule for the maintenance phase, if the ANC falls below 1.0 x 10⁹/L or the platelet count falls below 50 x 10⁹/L, the dose for the subsequent cycle must be reduced by one level (e.g., from 200 mg/m² to 150 mg/m², or from 150 mg/m² to 100 mg/m²). Treatment should be permanently discontinued if a patient cannot tolerate a dose of 100 mg/m²/day.[19]
IV Preparation and Administration
For intravenous use, the lyophilized powder must be carefully reconstituted and administered.
- Reconstitution: A 100 mg vial of Temozolomide powder is reconstituted with 41 mL of Sterile Water for Injection. The vial should be gently swirled, not shaken, to dissolve the powder. This yields a clear solution with a final concentration of 2.5 mg/mL.[19]
- Dilution and Infusion: The calculated volume of the reconstituted solution is withdrawn and transferred into an empty 250 mL infusion bag containing 0.9% Sodium Chloride injection only. The final solution is then administered as an intravenous infusion using a pump over a fixed period of 90 minutes.[19]
Table 3: Dosing Regimens for Approved Indications
Indication | Treatment Phase | Dose | Schedule |
---|
Newly Diagnosed GBM | Concomitant w/ RT | 75 mg/m²/day | Daily for 42-49 days |
Newly Diagnosed GBM | Maintenance (Cycle 1) | 150 mg/m²/day | Days 1-5 of 28-day cycle |
Newly Diagnosed GBM | Maintenance (Cycles 2-6) | 200 mg/m²/day | Days 1-5 of 28-day cycle |
Newly Diagnosed AA | Adjuvant (Cycle 1) | 150 mg/m²/day | Days 1-5 of 28-day cycle |
Newly Diagnosed AA | Adjuvant (Cycles 2-12) | 200 mg/m²/day | Days 1-5 of 28-day cycle |
Refractory AA | Initial (Cycle 1) | 150 mg/m²/day | Days 1-5 of 28-day cycle |
Refractory AA | Subsequent Cycles | 200 mg/m²/day | Days 1-5 of 28-day cycle |
Safety and Tolerability Profile
The clinical use of Temozolomide is associated with a well-defined spectrum of adverse events, ranging from common, manageable side effects to rare but serious toxicities that require careful monitoring and intervention.
A. Adverse Reactions
The safety profile of Temozolomide has been extensively characterized in clinical trials and postmarketing surveillance.
- Most Common Adverse Reactions: The most frequently reported side effects (occurring in ≥20% of patients) are largely related to the gastrointestinal and central nervous systems. These include alopecia (hair loss), fatigue, nausea, vomiting, headache, constipation, anorexia (loss of appetite), and convulsions.[17] Nausea and vomiting are particularly prevalent and often require management with prophylactic antiemetic medications.[5]
- Common Grade 3-4 Hematologic Abnormalities: The most clinically significant and dose-limiting toxicities of Temozolomide are hematologic. In clinical trials, the most common Grade 3 or 4 laboratory abnormalities (≥10% incidence) were lymphopenia (decrease in lymphocytes), thrombocytopenia (decrease in platelets), neutropenia (decrease in neutrophils), and leukopenia (decrease in total white blood cells).[22]
- Serious and Less Common Adverse Events:
- Hepatotoxicity: Cases of severe and sometimes fatal liver injury have been reported. It is mandatory to perform liver function tests at baseline, midway through the first cycle, prior to each subsequent cycle, and approximately two to four weeks after the final dose.[1]
- Pneumonia: Patients are at an increased risk of developing lung problems, including opportunistic infections. A particular concern is Pneumocystis pneumonia (PCP), especially when Temozolomide is given concurrently with radiotherapy and corticosteroids.[1]
- Severe Allergic/Skin Reactions: While mild to moderate rash is a common side effect, rare but life-threatening dermatologic reactions, such as Stevens-Johnson syndrome and toxic epidermal necrolysis, can occur.[17]
- Other Reported Events: A range of other adverse events have been observed, including dizziness, memory impairment, abnormal coordination, viral infections, and sores in the mouth and throat.[37]
B. Warnings, Precautions, and Contraindications
The FDA-approved prescribing information for Temozolomide includes several critical warnings and precautions to ensure patient safety.
- Contraindications: Temozolomide is strictly contraindicated in patients with a history of a serious hypersensitivity reaction (e.g., anaphylaxis) to Temozolomide or any of its excipients. Due to chemical similarity, it is also contraindicated in patients with a known hypersensitivity to its parent compound, dacarbazine (DTIC).[17]
- Myelosuppression: As detailed previously, bone marrow suppression is the primary dose-limiting toxicity. The risk is higher in geriatric patients and women.[1] Strict adherence to the blood count monitoring schedule is essential for safe administration.
- Secondary Malignancies: The very mechanism that makes Temozolomide an effective anticancer agent—its ability to induce DNA damage—also carries an inherent risk of long-term toxicity. By causing mutations in the DNA of healthy, rapidly dividing cells, such as hematopoietic stem cells in the bone marrow, Temozolomide can lead to the development of therapy-related secondary cancers. This is not an off-target effect but a direct, on-target consequence of its intended function. This creates an unavoidable risk-benefit calculation for every patient. Specifically, treatment with Temozolomide is associated with an increased risk of developing myelodysplastic syndrome (MDS) and secondary acute myeloid leukemia (AML).[1]
- Pneumocystis Pneumonia (PCP) Prophylaxis: Due to the risk of profound lymphopenia, especially during the concomitant phase of the Stupp protocol, prophylaxis against PCP is required for all patients receiving concurrent radiotherapy and Temozolomide. Prophylaxis should be continued for any patient who develops Grade 3 or 4 lymphopenia until it resolves.[1]
- Embryo-Fetal Toxicity: Temozolomide is teratogenic and can cause fetal harm. Therefore, it is critical to ensure that patients of reproductive potential use effective contraception. Females should use effective contraception during treatment and for at least 6 months after the final dose. Males should use condoms during treatment and for at least 3 months after the final dose, and should not donate semen during this period. Breastfeeding is not recommended during treatment and for at least one week after the last dose.[1]
- Use in Specific Populations: Caution should be exercised when administering Temozolomide to patients with severe renal or hepatic impairment, as its pharmacokinetics have not been formally studied in these populations.[5] While approved in the EU for children aged three and older with malignant glioma, its safety and efficacy have not been established for pediatric patients in the US for its primary indications of GBM and AA.[6]
C. Drug Interactions
While Temozolomide's non-enzymatic activation spares it from many common metabolic drug interactions, clinically significant pharmacodynamic interactions can occur. There are 277 drugs known to have potential interactions, with 55 classified as major.[60]
- Major Pharmacodynamic Interactions: The most critical interactions involve other agents that suppress or modulate the immune system.
- Live-Attenuated Vaccines: Co-administration of Temozolomide with live vaccines (e.g., measles, mumps, rubella, yellow fever) is generally not recommended. The drug-induced immunosuppression can lead to a diminished immune response to the vaccine and poses a risk of disseminated infection from the attenuated vaccine strain itself.[5]
- Cellular Immunotherapies: Similar concerns apply to advanced cellular therapies like CAR-T cells (e.g., axicabtagene ciloleucel), where the risk of infection and unpredictable immune effects is heightened.[53]
- Interactions with Myelosuppressive Agents: The risk of severe bone marrow suppression is increased when Temozolomide is combined with other drugs known to cause neutropenia or thrombocytopenia (e.g., deferiprone). Such combinations should be avoided or used with extreme caution and intensified hematologic monitoring.[53]
- Frequently Co-administered Drugs: Patients with high-grade gliomas are often on multiple concomitant medications, including antiepileptics (e.g., levetiracetam), corticosteroids for cerebral edema (dexamethasone), and antiemetics (e.g., ondansetron). While specific pharmacokinetic interactions with these agents are not well-defined, clinicians must be mindful of the potential for overlapping toxicities.[60]
Table 6: Common and Serious Adverse Reactions by Incidence
System Organ Class | Adverse Reaction | Frequency / Incidence (%) |
---|
Gastrointestinal | Nausea | 36-49% |
| Vomiting | 20-49% |
| Constipation | ≥20% |
| Anorexia | 19-61% |
Nervous System | Fatigue / Asthenia | 54-61% |
| Headache | ≥20% |
| Convulsions | ≥20% |
| Dizziness | ≥10% |
| Amnesia | ≥10% |
Skin & Subcutaneous Tissue | Alopecia (Hair Loss) | 55-69% |
| Rash | 19% |
Hematologic (Grade 3-4) | Lymphopenia | ≥10% |
| Thrombocytopenia | ≥10% |
| Neutropenia | ≥10% |
| Leukopenia | ≥10% |
Hepatobiliary | Hepatotoxicity (Severe) | Rare but reported |
Infections | Viral Infection | ≥10% |
| Pneumocystis Pneumonia (PCP) | Increased risk, especially with RT |
Neoplasms | Myelodysplastic Syndrome / Secondary Leukemia | Rare but reported |
The Molecular Basis of Temozolomide Resistance
The clinical success of Temozolomide is fundamentally constrained by the development of chemoresistance, a phenomenon observed in a majority of patients. This resistance is not driven by a single factor but is rather the result of a complex interplay of molecular pathways, with the DNA repair enzyme O⁶-methylguanine-DNA methyltransferase (MGMT) playing the central role.
A. The Central Role of O⁶-Methylguanine-DNA Methyltransferase (MGMT)
The expression level of the MGMT protein is the single most important determinant of a glioma's response to Temozolomide.
Mechanism of MGMT-Mediated Repair
As described previously, the primary cytotoxic lesion induced by Temozolomide is O6-methylguanine (O6-MeG). MGMT is a highly specialized DNA repair protein that functions as a "suicide enzyme" to counteract this specific type of damage.[61] It identifies the O6-MeG adduct on the DNA strand and directly transfers the aberrant methyl group from the guanine base to a cysteine residue within its own active site.[6] This action flawlessly restores the DNA to its original state but, in the process, the MGMT protein becomes irreversibly alkylated and is targeted for degradation. A cell's ability to withstand Temozolomide is therefore a direct function of its ability to synthesize new MGMT protein faster than it is consumed. Tumor cells with high levels of MGMT expression can efficiently repair the drug-induced lesions, leading to profound chemoresistance.[26]
MGMT Promoter Methylation as a Predictive Biomarker
In human cells, the expression of the MGMT gene is not typically regulated by mutations but rather by an epigenetic mechanism: methylation of its promoter region.[6] The promoter contains a high density of CpG dinucleotides (CpG islands) that, when methylated, act as a switch to turn off gene expression.
- Methylated Promoter ("MGMT-methylated"): When the CpG islands in the MGMT promoter are hypermethylated, the gene is transcriptionally silenced. As a result, the cell cannot produce MGMT protein. These tumors are unable to repair the critical O6-MeG lesions and are therefore highly sensitive to Temozolomide. In numerous clinical trials, MGMT promoter methylation has been established as the most powerful positive prognostic and predictive biomarker for GBM patients treated with Temozolomide, correlating strongly with improved progression-free and overall survival.[6]
- Unmethylated Promoter ("MGMT-unmethylated"): When the promoter is unmethylated, the MGMT gene is actively transcribed, leading to high levels of functional MGMT protein. These tumors can robustly repair DNA damage, exhibit strong resistance to Temozolomide, and patients with such tumors derive little to no survival benefit from the chemotherapy.[6]
Acquired Resistance and Changes in MGMT Status
Tumors are dynamic and can evolve under the selective pressure of therapy. There is mounting evidence that glioblastomas can acquire resistance to Temozolomide by altering their MGMT status. Studies have shown that tumors that are initially MGMT-methylated (and thus sensitive) can become unmethylated at the time of recurrence. This suggests that the chemotherapy eliminates the sensitive, methylated cells, allowing a small, pre-existing population of resistant, unmethylated cells to survive and repopulate the tumor, providing a clear molecular mechanism for acquired resistance.[61]
B. MGMT-Independent Resistance Mechanisms
While MGMT status is the dominant factor, it is not the sole determinant of response. A significant portion of patients with MGMT-methylated tumors still fail to respond to therapy, and over 50% of all GBM patients exhibit intrinsic resistance, indicating that other molecular pathways are critically involved.[26] This highlights that resistance is a complex network problem, not a single-gene issue.
The DNA Mismatch Repair (MMR) System
The MMR system is a crucial partner in mediating Temozolomide's cytotoxicity. Its role creates a critical dependency: for the drug to be effective in an MGMT-deficient cell, the MMR system must be fully functional. The MMR complex, which includes key proteins like MSH2 and MSH6, is responsible for recognizing the O6-MeG:T base mispair that forms during replication in the absence of MGMT repair.[28] It is this recognition that triggers the futile repair cycles and subsequent apoptosis.
If a tumor cell is not only MGMT-deficient but also has a co-existing deficiency in the MMR pathway, it enters a state of "damage tolerance." The cell cannot repair the O6-MeG lesion, but it also fails to recognize the resulting mismatch as a lethal error. The cell can therefore tolerate the DNA damage, bypass apoptosis, and continue to proliferate, rendering it resistant to Temozolomide even in the absence of MGMT.[28] This interplay explains why MGMT promoter methylation is a powerful but imperfect biomarker and underscores the importance of the MMR pathway as a key secondary mechanism of resistance.
The Base Excision Repair (BER) Pathway
The BER pathway is the primary mechanism for repairing the more frequent but less cytotoxic N7-MeG and N3-MeA lesions.[26] This pathway involves a cascade of enzymes, including the DNA glycosylase APNG and the sensor protein PARP (Poly(ADP-ribose) polymerase). While these lesions are not the primary drivers of cell death, hyperactivity of the BER pathway may contribute to overall cellular resilience and an enhanced ability to cope with generalized DNA damage, potentially lowering the threshold for apoptosis and contributing to a resistant phenotype.
Other Contributing Factors
The landscape of Temozolomide resistance is broad and includes several other cellular processes:
- Glioblastoma Stem Cells (GSCs): Tumors contain a subpopulation of cancer stem cells that are thought to be responsible for tumor initiation and recurrence. These GSCs often possess enhanced DNA repair capacity and other intrinsic survival mechanisms that make them highly resistant to chemotherapy and radiation.[29]
- Drug Efflux Pumps: Transmembrane proteins like P-glycoprotein (P-gp) can function as efflux pumps, actively transporting Temozolomide out of the tumor cell. This reduces the intracellular drug concentration and limits its ability to reach its DNA target, although its clinical significance in Temozolomide resistance is considered less prominent than that of the DNA repair pathways.[29]
- Signaling Pathways and Autophagy: Dysregulation of key intracellular signaling pathways, such as the PI3K/Akt pathway, can promote cell survival and contribute to resistance. Similarly, the process of autophagy, which normally degrades damaged cellular components, can be co-opted by cancer cells as a cytoprotective mechanism to survive the stress of chemotherapy.[27]
Future Perspectives and Strategies to Overcome Resistance
The profound challenge posed by Temozolomide resistance has spurred intensive research into novel strategies aimed at sensitizing tumors to its effects and improving clinical outcomes. The future of glioma therapy is evolving from a one-size-fits-all approach to a multi-pronged, personalized, and adaptive strategy that seeks to counteract the tumor's complex and dynamic escape mechanisms.
Targeting DNA Repair Pathways
Given that DNA repair is the principal mechanism of resistance, it is also the most logical therapeutic target.
- MGMT Inhibition and Depletion: A direct approach is to neutralize the MGMT enzyme in resistant tumors. Preclinical studies have used direct inhibitors like O6-benzylguanine (O6-BG) to deplete MGMT and sensitize cells to Temozolomide.[6] A more clinically advanced strategy involves using MGMT-depleting agents. For example, the proteasome inhibitor bortezomib, approved for multiple myeloma, has been shown to reduce MGMT protein levels in GBM cells. An ongoing clinical trial (BORTEM-17) investigating the combination of bortezomib and Temozolomide in recurrent GBM has shown promising interim results, particularly in MGMT-unmethylated patients.[62]
- PARP Inhibitors (PARPi): Targeting the BER pathway with PARP inhibitors is another promising strategy, particularly for tumors with MMR deficiencies. By inhibiting PARP, these drugs can prevent the repair of single-strand DNA breaks, leading to the accumulation of lethal double-strand breaks when combined with a DNA-damaging agent like Temozolomide. Several PARP inhibitors, including olaparib and veliparib, have the ability to cross the blood-brain barrier and are being actively investigated in combination with Temozolomide in numerous clinical trials for GBM.[44]
- BER Inhibition: Other agents that target the BER pathway, such as methoxyamine, work by blocking abasic (AP) sites that are created during the repair process. This has been shown to enhance the cytotoxicity of Temozolomide in preclinical models, irrespective of the tumor's MGMT status.[62]
Novel Drug Formulations and Analogs
Efforts are also underway to improve the drug itself or its delivery to the brain.
- Enhanced Delivery Systems: Overcoming the physical obstacle of the blood-brain barrier remains a key challenge. Advanced drug delivery platforms, such as nanoparticle-based systems, are being developed to encapsulate Temozolomide, improve its stability, and enhance its transport into the brain, thereby increasing its concentration at the tumor site.[63]
- Novel Analogs: Researchers are designing new chemical variants of Temozolomide to bypass known resistance mechanisms. One example is NEO212, a novel molecule that covalently fuses Temozolomide with perillyl alcohol, a natural compound with its own anti-glioma activity. This conjugate has demonstrated significantly greater anticancer activity than either parent molecule in preclinical studies.[6] Another modified drug, K-TMZ, was engineered to improve CNS penetration and showed promising results in mouse models.[12]
Combination with Other Therapeutic Modalities
The future of treatment likely lies in combining Temozolomide with other therapies to attack the tumor from multiple angles.
- Immunotherapy: Leveraging the known immunomodulatory effects of Temozolomide is a major area of focus. Combining the lymphodepleting effects of Temozolomide with immune checkpoint inhibitors (e.g., pembrolizumab, nivolumab) or other immune-stimulating agents aims to create a synergistic antitumor response. Multiple clinical trials are currently exploring these combinations.[1]
- Targeted Agents: As the molecular landscape of glioma becomes better understood, trials are combining Temozolomide with agents that target specific genetic alterations or dysregulated signaling pathways found in tumors. Examples include combinations with the nuclear export inhibitor selinexor and the CDK4/6 inhibitor abemaciclib.[44]
- Tumor-Treating Fields (TTFields): The Optune® device, which delivers low-intensity alternating electric fields to disrupt cancer cell division, is already established as a standard of care following the Stupp protocol. The ongoing EF-41/KEYNOTE D58 trial is testing its use even earlier, concurrently with the initial chemoradiation phase, in combination with both Temozolomide and pembrolizumab, representing a trimodal approach.[43]
Emerging and Preclinical Strategies
A host of innovative strategies are in earlier stages of development, reflecting a diversification of approaches to combat this difficult disease. These include targeting the unique metabolic vulnerabilities of glioma cells, manipulating the process of autophagy to promote cell death rather than survival, and using cutting-edge gene-editing technologies like CRISPR to directly disable resistance genes within the tumor.[63] This broad research portfolio signifies a paradigm shift away from a singular focus on cytotoxic chemotherapy and toward a more sophisticated, systems-level understanding of cancer biology, paving the way for a future of more effective and personalized treatments for patients with malignant glioma.
References
[1]
Works cited
- Temozolomide: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00853
- Temodar (temozolomide) FDA Approval History - Drugs.com, accessed July 16, 2025, https://www.drugs.com/history/temodar.html
- Temozolomide FAQs - UCSF Brain Tumor Center, accessed July 16, 2025, https://braintumorcenter.ucsf.edu/about-brain-tumors/treatment/medical-therapy/temozolomide-faqs
- en.wikipedia.org, accessed July 16, 2025, https://en.wikipedia.org/wiki/Temozolomide#:~:text=Temozolomide%2C%20sold%20under%20the%20brand,mouth%20or%20via%20intravenous%20infusion.
- Temozolomide (oral route) - Mayo Clinic, accessed July 16, 2025, https://www.mayoclinic.org/drugs-supplements/temozolomide-oral-route/description/drg-20066228
- Temozolomide - Wikipedia, accessed July 16, 2025, https://en.wikipedia.org/wiki/Temozolomide
- Temozolomide - CAS-Number 85622-93-1 - Order from Chemodex, accessed July 16, 2025, https://www.chemodex.com/products/temozolomide/
- www.medkoo.com, accessed July 16, 2025, https://www.medkoo.com/products/4761#:~:text=The%20chemical%20name%20of%20temozolomide,a%20molecular%20weight%20of%20194.15.
- Temozolomide | C6H6N6O2 | CID 5394 - PubChem, accessed July 16, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Temozolomide
- Temozolomide | 85622-93-1 - ChemicalBook, accessed July 16, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB2708199.htm
- Temozolomide (NSC 362856) | DNA Alkylator - MedchemExpress.com, accessed July 16, 2025, https://www.medchemexpress.com/Temozolomide.html
- Temozolomide - American Chemical Society, accessed July 16, 2025, https://www.acs.org/content/acs/en/molecule-of-the-week/archive/t/temozolomide.html
- Compound: TEMOZOLOMIDE (CHEMBL810) - ChEMBL - EMBL-EBI, accessed July 16, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL810
- Temozolomide | C6H6N6O2 - ChemSpider, accessed July 16, 2025, https://www.chemspider.com/Chemical-Structure.5201.html
- [Temozolomide (200 mg)] - CAS [85622-93-1] - USP Store, accessed July 16, 2025, https://store.usp.org/product/1643543
- Temozolomide - Sigma-Aldrich, accessed July 16, 2025, https://www.sigmaaldrich.com/US/en/product/mm/500609
- Temozolomide (Temodar): Uses, Side Effects, Interactions, Pictures, Warnings & Dosing, accessed July 16, 2025, https://www.webmd.com/drugs/2/drug-17512/temodar-oral/details
- 85622-93-1|3-Methyl-4-oxo-3,4-dihydroimidazo[5,1-d][1,2,3,5]tetrazine-8-carboxamide, accessed July 16, 2025, https://www.bldpharm.com/products/85622-93-1.html
- Temodar Dosage Guide - Drugs.com, accessed July 16, 2025, https://www.drugs.com/dosage/temodar.html
- Temozolomide Dosage Guide + Max Dose, Adjustments - Drugs.com, accessed July 16, 2025, https://www.drugs.com/dosage/temozolomide.html
- Indications and Usage (1.2) ------------- - accessdata.fda.gov - FDA, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/021029s036_037_038lbl.pdf
- Temodar Receives FDA Approval for New and Updated Indications and Doses Through the Project Renewal Program, accessed July 16, 2025, https://jhoponline.com/issue-archive/2023-issues/december-2023-vol-13-no-6/temodar-receives-fda-approval-for-new-and-updated-indications-and-doses-through-the-project-renewal-program
- TEMODAR® (temozolomide) capsules PHARMACIST: Dispense enclosed Patient Package Insert to each patient. PHARMACIST INFORMATION S - accessdata.fda.gov, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/21029s035lbl.pdf
- Generic Temodar Availability - Drugs.com, accessed July 16, 2025, https://www.drugs.com/availability/generic-temodar.html
- TEMODAR® (temozolomide) for injection PHARMACIST: Dispense enclosed Patient Package Insert to each patient. - accessdata.fda.gov, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022277s014lbl.pdf
- Temozolomide resistance in glioblastoma multiforme - PMC, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6150109/
- Acquired temozolomide resistance in MGMT-deficient glioblastoma cells is associated with regulation of DNA repair by DHC2 | Brain | Oxford Academic, accessed July 16, 2025, https://academic.oup.com/brain/article/142/8/2352/5538611
- An overview of glioblastoma multiforme and temozolomide resistance: can LC-MS-based proteomics reveal the fundamental mechanism of temozolomide resistance? - Frontiers, accessed July 16, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2023.1166207/full
- Temozolomide Resistance in Glioblastoma Cell Lines: Implication of MGMT, MMR, P-Glycoprotein and CD133 Expression | PLOS One, accessed July 16, 2025, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0140131
- Temozolomide | CAS 85622-93-1 | SCBT - Santa Cruz Biotechnology, accessed July 16, 2025, https://www.scbt.com/p/temozolomide-85622-93-1
- temozolomide | Cancer Care Ontario, accessed July 16, 2025, https://www.cancercareontario.ca/en/system/files_force/temozolomide.pdf?download=1
- 365-Glioblastoma temozolomide chemoradiation (part 1) - eviQ, accessed July 16, 2025, https://www.eviq.org.au/medical-oncology/neurological/glioma/365-glioblastoma-temozolomide-chemoradiation-part
- (PDF) Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma, accessed July 16, 2025, https://www.researchgate.net/publication/7976448_Radiotherapy_plus_Concomitant_and_Adjuvant_Temozolomide_for_Glioblastoma
- Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/15758009/
- Publisher's version PDF - Serval - UNIL, accessed July 16, 2025, https://serval.unil.ch/resource/serval:BIB_DEA725BDEE44.P001/REF.pdf
- Stupp protocol | Radiology Reference Article | Radiopaedia.org, accessed July 16, 2025, https://radiopaedia.org/articles/stupp-protocol
- TEMODAR (temozolomide) Label - accessdata.fda.gov, accessed July 16, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/021029s031lbl.pdf
- Glioblastoma temozolomide chemoradiation followed by temozolomide OVERVIEW - eviQ, accessed July 16, 2025, https://www.eviq.org.au/medical-oncology/neurological/glioma/3364-glioblastoma-temozolomide-chemoradiation-foll
- Chemotherapy for Low-Grade Gliomas: Lessons and Questions - Cancer Network, accessed July 16, 2025, https://www.cancernetwork.com/view/chemotherapy-low-grade-gliomas-lessons-and-questions
- Temozolomide based treatment in glioblastoma: 6 vs. 12 months - PMC - PubMed Central, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11240269/
- Temozolomide based treatment in glioblastoma: 6 vs. 12 months - Spandidos Publications, accessed July 16, 2025, https://www.spandidos-publications.com/10.3892/ol.2024.14551
- Temozolomide Completed Phase 2 Trials for Glioblastomas Treatment | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/drugs/DB00853/clinical_trials?conditions=DBCOND0052371&phase=2&purpose=treatment&status=completed
- Pivotal, Randomized, Open-label Study of Optune® (Tumor Treating Fields) Concomitant with RT & TMZ for the Treatment of Newly Diagnosed GBM - NCI, accessed July 16, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCT04471844
- Clinical Trials Using Temozolomide - NCI, accessed July 16, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/temozolomide
- Multicenter Phase II Trial of Temozolomide in Patients With Anaplastic Astrocytoma or Anaplastic Oligoastrocytoma at First Relapse - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/JCO.1999.17.9.2762
- Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse. Temodal Brain Tumor Group - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/10561351/
- Multicenter Phase II Trial of Temozolomide in Patients With Anaplastic Astrocytoma or Anaplastic Oligoastrocytoma at First Relapse - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/abs/10.1200/JCO.1999.17.9.2762
- Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse - Mayo Clinic, accessed July 16, 2025, https://mayoclinic.elsevierpure.com/en/publications/multicenter-phase-ii-trial-of-temozolomide-in-patients-with-anapl
- Adjuvant temozolomide improves survival in IDH-mutant anaplastic glioma - ecancer, accessed July 16, 2025, https://ecancer.org/en/video/12368-adjuvant-temozolomide-improves-survival-in-idh-mutant-anaplastic-glioma
- FDA Approves New Label for Temozolomide in Multiple Cancers - Targeted Oncology, accessed July 16, 2025, https://www.targetedonc.com/view/fda-approves-new-label-for-temozolomide-in-multiple-cancers
- Temozolomide (Temodar) Injection - Medical Clinical Policy Bulletins - Aetna, accessed July 16, 2025, https://www.aetna.com/cpb/medical/data/800_899/0876.html
- Pancreatic Neuroendocrine Tumor Completed Phase 1 / 2 Trials for Temozolomide (DB00853) | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/indications/DBCOND0036669/clinical_trials/DB00853?phase=1%2C2&status=completed
- Temodar (temozolomide) dosing, indications, interactions, adverse effects, and more, accessed July 16, 2025, https://reference.medscape.com/drug/temodar-temozolomide-342229
- temodar_pi.pdf - Merck.com, accessed July 16, 2025, https://www.merck.com/product/usa/pi_circulars/t/temodar_capsules/temodar_pi.pdf
- Temozolomide in the treatment of malignant brain tumors in children - Via Medica Journals, accessed July 16, 2025, https://journals.viamedica.pl/nowotwory_journal_of_oncology/article/download/53279/39926
- Temozolomide-related hematologic toxicity - PubMed, accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/23921765/
- Label: TEMOZOLOMIDE capsule - DailyMed, accessed July 16, 2025, https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=33d67bce-38ae-45fc-a858-d51167b0ecfd
- www.mayoclinic.org, accessed July 16, 2025, https://www.mayoclinic.org/drugs-supplements/temozolomide-oral-route/description/drg-20066228#:~:text=This%20medicine%20may%20cause%20serious,(eg%2C%20myeloid%20leukemia).
- Temozolomide: MedlinePlus Drug Information, accessed July 16, 2025, https://medlineplus.gov/druginfo/meds/a601250.html
- Temodar Interactions Checker - Drugs.com, accessed July 16, 2025, https://www.drugs.com/drug-interactions/temozolomide,temodar.html
- Mechanisms of temozolomide resistance in glioblastoma-a ..., accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8319838/
- A Review of Approaches to Potentiate the Activity of Temozolomide against Glioblastoma to Overcome Resistance - MDPI, accessed July 16, 2025, https://www.mdpi.com/1422-0067/25/6/3217
- Overcoming temozolomide resistance in glioma: recent advances ..., accessed July 16, 2025, https://pubmed.ncbi.nlm.nih.gov/40468460/
- Astrocytoma Clinical Trials - NCI, accessed July 16, 2025, https://www.cancer.gov/research/participate/clinical-trials/disease/astrocytoma?pn=1
- Glioblastoma Completed Phase Trials for Temozolomide (DB00853) | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/indications/DBCOND0002893/clinical_trials/DB00853?status=completed
- Astrocytoma of Brain Recruiting Phase Trials for Temozolomide (DB00853) - DrugBank, accessed July 16, 2025, https://go.drugbank.com/indications/DBCOND0106692/clinical_trials/DB00853?phase=&status=recruiting
- DIPG Terminated Phase 2 Trials for Temozolomide (DB00853) | DrugBank Online, accessed July 16, 2025, https://go.drugbank.com/indications/DBCOND0062673/clinical_trials/DB00853?phase=2&status=terminated
- Full article: Guideline Conformity to the Stupp Regimen in Patients with Newly Diagnosed Glioblastoma Multiforme in China - Taylor & Francis Online, accessed July 16, 2025, https://www.tandfonline.com/doi/full/10.2217/fon-2021-0435
- The Stupp regimen preceded by early post-surgery temozolomide versus the Stupp regimen alone in the treatment of patients with newly diagnosed glioblastoma multiforme (GBM). - ASCO Publications, accessed July 16, 2025, https://ascopubs.org/doi/10.1200/jco.2013.31.15_suppl.2022
- Study Details | Methoxyamine and Temozolomide in Treating Patients With Recurrent Glioblastoma | ClinicalTrials.gov, accessed July 16, 2025, https://clinicaltrials.gov/study/NCT02395692?term=AREA%5BBasicSearch%5D(AREA%5BConditionSearch%5D(%22Glioma%22)%20AND%20AREA%5BInterventionSearch%5D(%22Temozolomide%22))&rank=2
- Phase II trial of temozolomide (TMZ) plus irinotecan (CPT-11) in ..., accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2835159/
- Press Release: Clinical trial finds that adding the chemotherapy pill temozolomide to radiation therapy improves survival in adult patients with a slow-growing type of brain tumor - ECOG-ACRIN Cancer Research Group, accessed July 16, 2025, https://ecog-acrin.org/press-release-adding-the-chemotherapy-pill-temozolomide-to-radiation-therapy-improves-survival-in-adult-patients-with-glioma-a-slow-growing-type-of-brain-cancer/
- UCSD Astrocytoma Clinical Trials for 2025 — San Diego, accessed July 16, 2025, https://clinicaltrials.ucsd.edu/astrocytoma
- UCSF Astrocytoma Clinical Trials for 2025 — San Francisco Bay Area, accessed July 16, 2025, https://clinicaltrials.ucsf.edu/astrocytoma
- Phase III randomized study of radiation and temozolomide versus radiation and nitrosourea therapy for anaplastic astrocytoma: results of NRG Oncology RTOG 9813 - PubMed Central, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5463834/
- Real-World Evidence in Glioblastoma: Stupp's Regimen After a Decade - Frontiers, accessed July 16, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2020.00840/full
- Patterns of recurrence and outcomes of glioblastoma multiforme treated with chemoradiation and adjuvant temozolomide - SciELO Brasil, accessed July 16, 2025, https://www.scielo.br/j/clin/a/QDNQKTnTjTt5SvG8QpTSBkv/?lang=en
- Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse - MD Anderson Cancer Center, accessed July 16, 2025, https://mdanderson.elsevierpure.com/en/publications/multicenter-phase-ii-trial-of-temozolomide-in-patients-with-anapl
- Phase I Study of Temozolomide and Irinotecan for Recurrent Malignant Gliomas in Patients Receiving Enzyme-Inducing Antiepileptic Drugs - PubMed Central, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4802002/
- BC Cancer Protocol Summary for Therapy for Malignant Brain Tumours using Temozolomide, accessed July 16, 2025, http://www.bccancer.bc.ca/chemotherapy-protocols-site/Documents/Neuro-Oncology/CNTEMOZ_Protocol.pdf
- TA23 Brain cancer - temozolomide: HTA Report - NICE, accessed July 16, 2025, https://www.nice.org.uk/guidance/ta23/documents/ta23-brain-cancer-temozolomide-hta-report2
- Do Extent of Resection and Tumor Volume affect the Overall Survival of Anaplastic Astrocytoma? A Retrospective Study from a Single Center, accessed July 16, 2025, https://oamjms.eu/index.php/mjms/article/view/10697
- EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma - ZORA (Zurich Open Repository and Archive), accessed July 16, 2025, https://www.zora.uzh.ch/id/eprint/98103/7/Sammelmappe1.pdf
- A phase II study of temozolomide in patients with newly diagnosed supratentorial malignant glioma before radiation therapy, accessed July 16, 2025, https://academic.oup.com/neuro-oncology/article-pdf/4/4/261/13065876/NO_4_4-04p261.pdf
- Treatment of Glioblastoma with Intravenous Taurolidine. First Clinical Experience - Anticancer Research, accessed July 16, 2025, https://ar.iiarjournals.org/content/anticanres/24/2C/1143.full-text.pdf
- Temozolomide Salvage Chemotherapy for Recurrent Anaplastic Oligodendroglioma and Oligo-Astrocytoma - PMC, accessed July 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3921276/
- Temozolomide in Treating Patients With Newly Diagnosed Anaplastic Oligodendroglioma or Mixed Oligoastrocytoma | ClinicalTrials.gov, accessed July 16, 2025, https://clinicaltrials.gov/study/NCT00400816
- Study Details | Temozolomide in Patients With Newly Diagnosed Anaplastic Oligodendroglioma and Mixed Oligoastrocytoma | ClinicalTrials.gov, accessed July 16, 2025, https://www.clinicaltrials.gov/study/NCT00362570