Fact-checked by Grok 2 weeks ago

Oxaliplatin

Oxaliplatin is a third-generation platinum-based antineoplastic agent used primarily in combination with infusional fluorouracil and leucovorin for the treatment of colorectal cancer, including adjuvant therapy following resection of stage III colon cancer and treatment of advanced or metastatic disease. Marketed under the brand name Eloxatin by Sanofi-Aventis, it was initially approved by the U.S. Food and Drug Administration (FDA) in 2002 for metastatic colorectal cancer, with expanded approval in 2004 for adjuvant use. As a chemotherapeutic drug, oxaliplatin exerts its cytotoxic effects through the formation of DNA crosslinks, which inhibit DNA replication and transcription in cancer cells, leading to cell cycle arrest and apoptosis; its activity is cell-cycle nonspecific. Developed in the 1970s and 1980s as an alternative to earlier platinum compounds like cisplatin, which exhibited significant nephrotoxicity and limited efficacy against colorectal tumors, oxaliplatin was first investigated in phase I trials by Mathé et al. in 1986, demonstrating tolerability at doses up to 67 mg/m² without renal impairment. Subsequent studies, including phase II trials by Machover et al. (1996) and Diaz-Rubio et al. (1998), confirmed its activity in advanced colorectal cancer, paving the way for phase III trials such as the de Gramont et al. study (2000), which established the efficacy of the FOLFOX regimen (oxaliplatin, 5-fluorouracil, and leucovorin) in improving response rates and survival. Administered intravenously at a standard dose of 85 mg/m² over 2 hours every two weeks as part of a 14-day cycle, oxaliplatin is typically given for up to 12 cycles in adjuvant settings or until disease progression in advanced cases, with dose adjustments based on toxicity. While effective, oxaliplatin is associated with notable toxicities, including peripheral sensory neuropathy—often acute and cold-induced, affecting up to 90% of patients—which is often reversible but may persist in approximately 30% of cases at 18 months post-treatment (21% grade 1, 10% grade 2); other common adverse effects encompass myelosuppression, nausea, vomiting, fatigue, and hypersensitivity reactions that can be severe or fatal. Additional risks include hepatotoxicity, pulmonary fibrosis, and QT interval prolongation, necessitating careful monitoring of blood counts, liver function, and cardiac activity during therapy. Contraindicated in patients with known hypersensitivity to platinum compounds, oxaliplatin's role in oncology has evolved with molecular targeted therapies, yet it remains a cornerstone in colorectal cancer management due to its synergistic effects in combination regimens.

Chemistry

Molecular Structure

Oxaliplatin is a third-generation platinum-based anticancer agent with the chemical formula [Pt(ox)(DACH)], where "ox" denotes the bidentate oxalate ligand (C₂O₄²⁻) and "DACH" refers to (1R,2R)-1,2-diaminocyclohexane, a bidentate amine ligand. At the core of its structure is a central platinum(II) atom coordinated to four donor atoms in a square planar geometry, typical of d⁸ transition metal complexes: two oxygen atoms from the oxalate ligand and two nitrogen atoms from the DACH ligand. The oxalate acts as a chelating leaving group, bridging the platinum via its two oxygen atoms, while the DACH ligand chelates through its amine groups in a trans configuration, with both nitrogen donors occupying adjacent positions in the coordination plane. This arrangement results in a neutral, overall mononuclear complex with the molecular formula C₈H₁₄N₂O₄Pt and a molecular weight of 397.29 g/mol. In comparison to first-generation cisplatin ([Pt(NH₃)₂Cl₂]) and second-generation carboplatin ([Pt(NH₃)₂(cyclobutanedicarboxylate)]), oxaliplatin features a bulkier, bidentate DACH carrier ligand instead of two monodentate ammonia groups, introducing stereochemistry via two chiral centers at the 1 and 2 positions of the cyclohexane ring in the (1R,2R)-enantiomer. This chiral, non-leaving DACH enhances ligand stability and steric hindrance around the platinum center, differing from the symmetric, achiral ammonia ligands in cisplatin and carboplatin, while the oxalate leaving group provides greater inertness than cisplatin's chlorides but higher lability than carboplatin's bidentate cyclobutanedicarboxylate. Structural representations of oxaliplatin typically include 2D depictions showing the square planar coordination with the DACH ring in a chair conformation and the oxalate as a five-membered chelate ring, alongside 3D models that highlight the equatorial positioning of the amine substituents and the overall molecular asymmetry. These features contribute to its distinct reactivity profile, as the bulky DACH ligand influences substitution rates at the platinum center by modulating access to the leaving group, promoting selective ligand exchange over the more reactive profiles of earlier platinum drugs.

Physical and Chemical Properties

Oxaliplatin appears as a white to off-white crystalline powder. Its molecular formula is C₈H₁₄N₂O₄Pt, with a molecular weight of 397.29 g/mol. The compound has a melting point of approximately 198 °C, at which it decomposes. Oxaliplatin exhibits low solubility in water, at about 6 mg/mL, rendering it slightly soluble; it is very slightly soluble in methanol and practically insoluble in ethanol and acetone. This limited aqueous solubility stems from its molecular structure, which includes a lipophilic cyclohexanediamine ligand. In pharmaceutical formulations, the injection solution is prepared in water with stabilizers such as lactate to enhance handling. The stability of oxaliplatin is sensitive to light, temperature, and pH conditions. Concentrated solutions must be protected from light, and storage is recommended at controlled room temperature (20–25 °C), with excursions permitted to 15–30 °C; freezing is prohibited to avoid degradation. In solution, oxaliplatin undergoes pH-dependent hydrolysis, showing instability in chloride-containing or alkaline media but greater stability in acidic glucose solutions, such as 5% dextrose at pH around 3.5–5.5. Diluted solutions remain stable for up to 24 hours under refrigeration (2–8 °C) or 6 hours at room temperature. Pharmaceutical-grade oxaliplatin adheres to United States Pharmacopeia (USP) standards, requiring a purity of not less than 98.0% and not more than 102.0% on the dried basis to ensure efficacy and safety in clinical use.

Pharmacology

Mechanism of Action

Oxaliplatin, a third-generation platinum-based chemotherapeutic agent, primarily exerts its anticancer effects through the formation of DNA adducts following intracellular activation. Upon entering cells via passive diffusion or copper transporter 1 (CTR1), oxaliplatin undergoes aquation in the low-chloride intracellular environment (~4 mM Cl⁻ compared to ~100-140 mM in plasma), where the labile oxalate ligand is displaced by water molecules. This non-enzymatic process generates reactive platinum species, such as the monoaqua [Pt(H₂O)(OH)(DACH)]⁺ and diaqua [Pt(H₂O)₂(DACH)]²⁺ complexes, with the simplified aquation reaction represented as: [\ce{Pt(ox)(DACH)}] + \ce{H2O} \rightarrow [\ce{Pt(H2O)(OH)(DACH)}]+ + \ce{oxalate} These aquated species are electrophilic and capable of binding to nucleophilic sites on DNA, with the rate of aquation being pH- and chloride-dependent, accelerating under physiological intracellular conditions. The activated platinum complexes predominantly form intrastrand crosslinks on DNA, with approximately 55-60% being 1,2-d(GpG) adducts involving adjacent guanines, alongside lesser proportions of 1,2-d(ApG) and 1,3-d(GpNpG) intrastrand links, as well as interstrand crosslinks (~2-5%) and DNA-protein crosslinks. These adducts distort the DNA helix, bending it by about 30-40° and unwinding it, which inhibits DNA replication and transcription by blocking polymerases and helicases. The bulky DACH ligand enhances the conformational rigidity and hydrophobicity of these adducts compared to those formed by cisplatin, leading to greater interference with DNA processing enzymes. The DNA damage triggers cellular responses, including activation of the nucleotide excision repair (NER) pathway, which attempts to remove the bulky adducts but can become overwhelmed, resulting in persistent DNA lesions and replication fork stalling. This activates ataxia-telangiectasia mutated (ATM)/ataxia-telangiectasia and Rad3-related (ATR) kinases, leading to p53 stabilization and phosphorylation, upregulation of p21, and subsequent G2/M cell cycle arrest. Ultimately, unresolved damage induces apoptosis through mitochondrial pathways, involving Bax translocation, cytochrome c release, and caspase-3 activation, with p53-dependent mechanisms amplifying the response in tumor cells. Compared to cisplatin, oxaliplatin's DACH ligand confers key advantages, including reduced reactivity toward sulfur-containing nucleophiles like glutathione and other thiols due to steric hindrance, which minimizes extracellular inactivation and lowers nephrotoxicity while preserving intracellular efficacy. Additionally, the stereospecific (1R,2R)-trans-DACH isomer in oxaliplatin produces adducts that evade mismatch repair (MMR) proteins more effectively than cisplatin's cis-diammine ligands, bypassing resistance mechanisms in MMR-deficient tumors and reducing cross-resistance. These properties contribute to oxaliplatin's distinct cytotoxicity profile despite forming fewer DNA adducts overall.

Pharmacokinetics

Oxaliplatin is administered exclusively via intravenous infusion, with no oral bioavailability due to its chemical instability and poor gastrointestinal absorption. Typical regimens involve doses of 85 mg/m² over 2 hours every 2 weeks or 130 mg/m² every 3 weeks, often in combination with other agents. Following intravenous administration, oxaliplatin exhibits rapid distribution with a large volume of distribution of approximately 440 L, indicating extensive tissue penetration. About 85% of the dose is distributed to tissues or excreted shortly after infusion, while plasma protein binding exceeds 90%, primarily to albumin and gamma-globulins. Oxaliplatin minimally crosses the blood-brain barrier, with cerebrospinal fluid penetration of only about 1.6% relative to plasma concentrations. Metabolism occurs through rapid, non-enzymatic biotransformation in plasma and tissues, yielding up to 17 platinum-containing derivatives, including active cytotoxic species such as monochloro- and diaquo-DACH platinum complexes; hepatic cytochrome P450 enzymes are not involved. One key metabolite is Pt(DACH)Cl₂. Elimination is primarily renal, with 54% of administered platinum recovered unchanged in urine within 5 days; fecal excretion accounts for about 2%. Pharmacokinetics follow a triphasic profile in plasma ultrafiltrate, with half-lives of 0.43 hours (alpha), 16.8 hours (beta), and 392 hours (terminal gamma phase). Total plasma clearance ranges from 10 to 17 L/h, exceeding glomerular filtration rate and correlating with renal function. Pharmacokinetics are unaffected by age, sex, or hepatic impairment, but renal dysfunction significantly increases exposure: unbound platinum AUC rises by 40%, 95%, and 342% in mild, moderate, and severe impairment, respectively, compared to normal renal function. Dose adjustments are recommended for severe renal impairment (creatinine clearance <30 mL/min).

Medical Uses

Approved Indications

Oxaliplatin is approved by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) primarily for the treatment of colorectal cancer in combination with infusional 5-fluorouracil (5-FU) and leucovorin (LV), known as the FOLFOX regimen. This includes its use as a first-line therapy for advanced (metastatic) colorectal cancer, where it has demonstrated improved response rates and progression-free survival compared to 5-FU/LV alone in pivotal trials. For adjuvant therapy, oxaliplatin is indicated in patients with stage III colon cancer following complete resection of the primary tumor, administered with 5-FU/LV for up to 6 months to reduce the risk of recurrence. The MOSAIC trial, a landmark phase 3 study, showed that adding oxaliplatin to 5-FU/LV reduced the relative risk of relapse or death by 23% at 3 years, with a disease-free survival rate of 78.2% versus 72.9% in the control arm. In certain regions, such as Japan, oxaliplatin has additional approvals for advanced gastric cancer, often in combination regimens like SOX (S-1 plus oxaliplatin). For pancreatic cancer, oxaliplatin is incorporated into FDA-approved regimens like NALIRIFOX (nanoliposomal irinotecan, 5-FU, leucovorin, and oxaliplatin) for first-line treatment of metastatic disease, though its standalone approval remains limited to colorectal indications. Off-label uses are emerging for other malignancies, including ovarian cancer and non-small cell lung cancer (NSCLC), based on ongoing clinical investigations, but these are not yet formally approved.

Dosage and Administration

Oxaliplatin is administered intravenously as part of combination chemotherapy regimens for colorectal cancer. In the FOLFOX regimen, the standard dose is 85 mg/m² infused over 2 hours on day 1 of a 14-day cycle, in combination with leucovorin and fluorouracil. For adjuvant therapy in stage III colon cancer, an alternative regimen is XELOX (or CAPOX), where oxaliplatin is given at 130 mg/m² over 2 hours on day 1 of a 21-day cycle, combined with oral capecitabine. Preparation of oxaliplatin requires reconstitution of the lyophilized powder with sterile water for injection, followed by dilution in 250 to 500 mL of 5% dextrose in water (D5W); chloride-containing solutions must be avoided to prevent drug inactivation. The diluted solution should be inspected for particulates and discoloration, and protected from light during handling. Administration occurs via a 2-hour intravenous infusion through a dedicated line, with concurrent infusion of leucovorin in a separate bag if applicable; aluminum-containing equipment must be avoided to prevent degradation of the platinum compound. Premedication with antiemetics is recommended to manage potential nausea and vomiting. Patients should avoid cold exposure, including cold drinks and environments, during and shortly after infusion to minimize exacerbation of acute peripheral neuropathy symptoms. Dose adjustments are necessary for toxicity management. For persistent grade 2 peripheral sensory neuropathy in the adjuvant setting, the dose may be reduced to 75 mg/m² in the FOLFOX regimen; in advanced disease, reduce to 65 mg/m². For grade 3 or higher, discontinuation is considered. In the XELOX regimen, similar reductions apply, typically to 100 mg/m² initially for grade 2 neuropathy, with further adjustments or omission for higher grades. For severe renal impairment (creatinine clearance <30 mL/min), the initial dose should be reduced to 65 mg/m²; there is no contraindication based on renal function. Treatment duration in metastatic colorectal cancer is continued until disease progression or unacceptable toxicity, often up to 12 cycles. In the adjuvant setting for stage III colon cancer, therapy is limited to 12 cycles (approximately 6 months) with either FOLFOX or XELOX.

Adverse Effects

Peripheral Neuropathy

Peripheral neuropathy is a hallmark and often dose-limiting adverse effect of oxaliplatin chemotherapy, presenting in acute and chronic forms that significantly impact patient quality of life. The incidence of acute oxaliplatin-induced peripheral neuropathy (OIPN) ranges from 4% to 98% across studies, commonly affecting 85-96% of patients within hours of infusion and typically resolving within 7 days. Chronic OIPN develops cumulatively, with rates of 40-93% at doses exceeding 780-850 mg/m², and may persist for months to years even after treatment cessation. At high cumulative doses around 1020 mg/m², severe (grade 3) neurotoxicity occurs in 12-18% of patients. The pathophysiology of OIPN involves platinum accumulation in the dorsal root ganglia (DRG), leading to axonal degeneration and sensory neuron damage. Oxaliplatin and its oxalate metabolites induce oxidative stress and mitochondrial dysfunction in DRG neurons, impairing axonal transport and causing neuronopathy. A key mechanism is ion channel dysfunction, particularly the sensitization of transient receptor potential ankyrin 1 (TRPA1) channels via inhibition of prolyl hydroxylase and intracellular pH acidification, which exacerbates cold-induced hyperexcitability in peripheral nerves. These changes contribute to both acute transient effects and chronic persistent damage. Symptoms of acute OIPN primarily include cold-triggered distal paresthesia and dysesthesia in the hands, feet, perioral region, and jaw, often accompanied by tightness or spasms in the jaw, voice changes, and muscle cramps. Chronic OIPN manifests as persistent sensory loss, numbness, tingling, and proprioceptive deficits, leading to functional impairments such as difficulty writing, handling utensils, or maintaining balance. These symptoms can worsen during or shortly after infusion and may exhibit a "coasting" effect, intensifying for 2-3 months post-treatment in up to 60% of cases. Risk factors for developing severe OIPN include cumulative oxaliplatin doses greater than 800 mg/m², which markedly increase the likelihood of chronic symptoms. Patient-related factors encompass older age, female sex, diabetes, obesity (higher BMI), anemia, hypomagnesemia, alcohol consumption, and preexisting neuropathy. Genetic predispositions, such as variants in the AGXT gene (involved in oxalate metabolism), along with polymorphisms in GSTP1, OCT2, and SCN9A, have been associated with heightened susceptibility. Monitoring of OIPN relies on standardized grading using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE), which assesses sensory symptoms like paresthesia and interference with daily activities on a scale from grade 1 (asymptomatic) to grade 4 (disabling). Patients are evaluated before each cycle, with dose reduction or discontinuation recommended for grade 3 (severe symptoms limiting self-care) or higher to prevent irreversible damage. Regular clinical assessment, including patient-reported outcomes, helps track progression and guide management.

Other Adverse Effects

Oxaliplatin treatment is associated with significant hematologic toxicities, primarily manifesting as myelosuppression, which includes neutropenia and thrombocytopenia. Neutropenia occurs in approximately 41% of patients as grade 3 or 4 events in adjuvant settings and 53% in advanced colorectal cancer, with a nadir typically reached between days 7 and 14 post-administration. Thrombocytopenia is reported at rates of 2-5% for severe grades, and these effects are generally reversible upon dose adjustment or interruption. Regular monitoring of complete blood counts (CBC) is recommended to manage myelosuppression and prevent complications such as febrile neutropenia. Gastrointestinal adverse effects are common and often moderate in severity. Nausea affects 71-74% of patients overall, with vomiting in 41-47%, and grade 3 or 4 events occurring in 4-6%; these are typically managed with antiemetics. Diarrhea is observed in 56% of cases, with severe grades in 11-12%, while mucositis or stomatitis impacts 38-42%, rarely exceeding grade 3 (0-3%). Incidence rates from pivotal clinical trials, such as those in the OPTIMOX studies, align with these figures, highlighting the need for supportive care to maintain treatment continuity. Other non-neurological effects include reactions, affecting about 10% of patients overall and 3% severely, with being rare but potentially life-threatening, often occurring after multiple cycles. is less frequent than with , primarily reported in as transient or rather than permanent damage. imbalances, particularly hypomagnesemia, arise due to renal dysfunction and may persist post-treatment, necessitating periodic and supplementation. Hepatotoxicity is another potential adverse effect, manifesting as elevated liver enzymes (incidence ~10-20% for grade 3/4) or more specifically sinusoidal obstruction syndrome and vascular injury, reported in up to 19-52% of patients in some studies, which may affect surgical outcomes in colorectal cancer. Pulmonary fibrosis is rare (<1% incidence) but can be severe or fatal, typically occurring after prolonged exposure. Additionally, oxaliplatin may cause QT interval prolongation and ventricular arrhythmias, including fatal torsade de pointes; electrocardiogram monitoring is recommended, particularly in patients with cardiac risk factors or concomitant QT-prolonging drugs.

Contraindications and Precautions

Contraindications

Oxaliplatin is contraindicated in patients with a known hypersensitivity to the drug, other platinum-based compounds, or any of its excipients, as administration can provoke severe and potentially fatal reactions such as anaphylaxis, bronchospasm, or hypotension occurring within minutes of infusion. In such cases, the drug must be permanently discontinued, and appropriate resuscitative measures initiated. According to some international guidelines (e.g., UK SmPC and Canadian Product Monograph), severe renal impairment ( clearance <30 mL/min) is an absolute due to risk of accumulation and . However, per US FDA guidelines, it is a precaution requiring dose to 65 mg/m². Per the Canadian Product Monograph, oxaliplatin is contraindicated during due to demonstrated teratogenic and embryotoxic effects in , with potential for fetal harm or lethality. The US FDA states it can cause fetal harm and advises effective contraception for females of reproductive potential during treatment and for 9 months after the final dose, and for males during treatment and for 6 months after. Relative contraindications include pre-existing peripheral sensory neuropathy of grade 2 or higher with functional impairment, as oxaliplatin can exacerbate , potentially leading to irreversible damage. is contraindicated per and Canadian guidelines to excretion into breast milk and risks to the infant; the FDA advises against breastfeeding during treatment and for 3 months after the final dose. Patients with unresolved gastrointestinal issues require caution and to potential for worsening ileus, obstruction, or dehydration from treatment-related nausea and diarrhea. Precautions are advised in elderly patients, who experience higher rates of severe toxicity, including grade 3/4 events like diarrhea and neuropathy, necessitating close monitoring and potential dose adjustments. Prior history of neuropathy, even if resolved, requires careful , as cumulative increases recurrence . These restrictions stem from oxaliplatin's pharmacokinetic profile, involving renal elimination and platinum-DNA formation that amplifies and neurotoxic risks in vulnerable populations.

Drug Interactions

Oxaliplatin is primarily eliminated through the kidneys, and coadministration with nephrotoxic agents can decrease its clearance, leading to additive renal toxicity. Examples include aminoglycosides and nonsteroidal anti-inflammatory drugs (NSAIDs), for which avoidance or close monitoring of creatinine clearance is recommended. Oxaliplatin can prolong the QT interval, and concurrent use with other QT-prolonging drugs should be avoided to prevent ventricular arrhythmias. This includes 5-HT3 receptor antagonists such as ondansetron, which may enhance this risk when used as antiemetics during chemotherapy. When combined with and leucovorin, oxaliplatin has no significant pharmacokinetic interaction at standard doses of 85 mg/m² every 2 weeks, though higher doses of 130 mg/m² every 3 weeks may increase plasma concentrations by approximately 20%. In contrast, coadministration with taxanes such as can increase the risk of myelosuppression and ; taxanes should be administered to oxaliplatin if combined. Prolonged prothrombin time and international normalized ratio (INR), occasionally associated with hemorrhage, have been reported in patients receiving oxaliplatin with fluorouracil/leucovorin and oral anticoagulants; more frequent monitoring of these parameters is advised. Oxaliplatin undergoes nonenzymatic biotransformation and does not inhibit cytochrome P450 isoenzymes, so no major interactions via CYP450 pathways are anticipated. A history of alcohol consumption may predispose patients to worsened peripheral neuropathy due to preexisting nerve damage.

History

Development

Oxaliplatin was first synthesized in 1976 by Japanese chemist Yoshinori Kidani at Nagoya City University as part of research to create platinum-based anticancer agents with reduced toxicity and broader activity compared to cisplatin. The compound, featuring a 1,2-diaminocyclohexane (DACH) carrier ligand and an oxalate leaving group, was later licensed to the Swiss pharmaceutical company Debiopharm for clinical development. In preclinical evaluations, oxaliplatin exhibited potent antitumor activity in the L1210 murine leukemia model, where it outperformed other platinum analogs. It was classified as a third-generation platinum agent due to its effectiveness against cisplatin-resistant cell lines, attributed to the non-cross-resistant DACH ligand that forms distinct DNA adducts and induces apoptosis through unique pathways. These properties highlighted its potential for treating tumors unresponsive to earlier platinums, prompting selection for advanced testing. Early clinical development began in the late 1980s and 1990s with phase I trials to determine safety and dosing. A 1990 phase I study escalated doses up to 200 mg/m² every three weeks, identifying peripheral sensory neuropathy as the dose-limiting toxicity and establishing a maximum tolerated dose (MTD) of 130–135 mg/m². Subsequent phase II trials confirmed efficacy in advanced colorectal cancer; for instance, a 1993 EORTC Gastrointestinal Tract Cancer Cooperative Group study administered oxaliplatin via 5-day circadian-modulated infusion (up to 200 mg/m² per course) to 30 pretreated patients, yielding a 10% objective response rate with manageable toxicity, primarily diarrhea and neuropathy. A key milestone occurred in a 1996-initiated phase III multicenter randomized trial, which compared oxaliplatin (85 mg/m²) added to chronomodulated 5-fluorouracil (5-FU) and leucovorin (LV) versus 5-FU/LV alone as first-line therapy for metastatic colorectal cancer. The combination achieved superior objective response rates (50.7% versus 22.3%) and progression-free survival (9.0 versus 6.2 months), establishing oxaliplatin's role in combination regimens while underscoring its non-overlapping toxicity profile with 5-FU.

Regulatory Approvals

Oxaliplatin received its first approval in Europe in France in April 1996 for second-line treatment of metastatic colorectal cancer in combination with infusional 5-fluorouracil and leucovorin, with the indication extended to first-line treatment in April 1998. It was subsequently extended through mutual recognition procedures to other member states for second-line therapy in advanced colorectal cancer. In the United States, the Food and Drug Administration (FDA) granted initial approval to oxaliplatin (as Eloxatin) on August 9, 2002, for the treatment of advanced colorectal cancer in combination with infusional 5-fluorouracil and leucovorin, specifically for patients whose disease had progressed following treatment with irinotecan-based therapy. The indication was expanded on January 9, 2004, to include first-line treatment of advanced colorectal cancer in combination with the same regimen. Further expansion occurred in August 2004 for adjuvant treatment of stage III colon cancer following complete resection, based on demonstrated disease-free survival benefits in pivotal trials like the MOSAIC study. Approvals in other regions followed a similar timeline. Health Canada approved oxaliplatin in 2000 for metastatic under special access provisions, with full notice of compliance granted in 2007 for broader use in therapies. In Japan, the Ministry of Health, Labour and Welfare approved oxaliplatin in March 2005 for advanced or recurrent , marking it as the first platinum agent for this indication in the country. Oxaliplatin was added to the Organization's Model List of in 2005, recognizing its role in standard treatment protocols globally. Subsequent label expansions incorporated combination regimens supported by phase III trials. For instance, the NO16966 trial demonstrated the efficacy of oxaliplatin-based regimens (XELOX or FOLFOX) with bevacizumab in first-line metastatic colorectal cancer, leading to FDA and EMA approvals for these combinations in 2006 and 2007, respectively, improving progression-free survival without compromising quality of life. Further expansions included approval for use in combination with capecitabine (XELOX regimen) by the FDA in 2007 and EMA shortly after, as well as additional indications such as advanced pancreatic cancer (FDA 2011, EMA 2017) and gastric cancer in select regions post-2015. It remains on the WHO Model List of Essential Medicines as of the 23rd edition (2023). Post-marketing prompted updates to labeling. In 2007, the FDA revised the Eloxatin prescribing to strengthen warnings on severe peripheral sensory neuropathy, emphasizing dose reductions or discontinuation based on cumulative dose and symptom severity, following reports of persistent effects in up to 20% of patients. Similar updates were implemented by the , highlighting the of cold-induced acute neuropathy and long-term sensory deficits.

Society and Culture

Brand Names and Formulations

Oxaliplatin is commercially available under the primary brand name Eloxatin, developed and marketed by Sanofi. Eloxatin was discontinued in the United States in 2024. Following the expiration of patents, generic versions of oxaliplatin have been produced by multiple pharmaceutical companies, including Teva Pharmaceuticals, Hospira (now part of Pfizer), Accord Healthcare, and Actavis (now part of Teva). Examples of generic trade names include Oxaliplat and various oxaliplatin injections offered by these manufacturers. The drug is formulated exclusively for intravenous administration, with no oral dosage forms available. Common presentations include lyophilized powder for injection in mg and 100 mg single-use vials, which require reconstitution prior to dilution and infusion. Ready-to-use injection solutions are also available as mg/10 mL (5 mg/mL) or 100 mg/20 mL (5 mg/mL) clear, colorless concentrates in single-dose vials. Combination packs with 5-fluorouracil are not standard formulations for oxaliplatin.

Availability and Cost

Oxaliplatin has been widely available as a generic medication since the expiration of key patents in the United States in 2013 and in the European Union around the same period, enabling multiple manufacturers to produce and distribute affordable versions globally. In developed countries such as the US and those in the EU, it is readily accessible through standard pharmaceutical supply chains, with generics dominating the market and ensuring broad availability in hospitals and oncology centers, though shortages of certain presentations have been reported as of 2025, including national backorders in the US. The drug is also included on the World Health Organization's Model List of Essential Medicines, facilitating its procurement and use in low- and middle-income countries (LMICs) for essential cancer treatments, though distribution challenges persist in remote or under-resourced areas. The cost of generic oxaliplatin in the US has significantly decreased following patent expiry, with prices typically ranging from $20 to $100 per treatment cycle (as of 2025), depending on dosage (e.g., 85-170 mg/m² every two weeks) and vial sizes. The introduction of generics has driven substantial price reductions of 70-90% compared to branded versions, improving overall affordability and enabling wider adoption in treatment regimens worldwide. As of 2025, it is included in various national formularies, such as the US Veterans Affairs National Formulary and Singapore's National Drug Formulary, supporting its integration into public health systems.