Fact-checked by Grok 2 weeks ago

FOLFIRI

FOLFIRI is a combination chemotherapy regimen consisting of (leucovorin), (5-FU), and , primarily used to treat advanced or metastatic . This regimen enhances the efficacy of by incorporating , which stabilizes the fluorouracil-thymidylate synthase complex to inhibit , while , a I inhibitor, causes DNA damage in rapidly dividing cancer cells. Developed as an evolution of earlier fluorouracil-leucovorin combinations, FOLFIRI incorporates to improve response rates and survival outcomes in patients with metastatic disease. The regimen follows a biweekly schedule, typically administered intravenously on day 1 of a 14-day : at 180 mg/m² over 90 minutes, concurrent with at 400 mg/m² over 120 minutes, followed by a bolus of 400 mg/m² and a continuous of 2400 mg/m² over 46 hours. Treatment is often continued for up to 12 s, or about 6 months, depending on patient response and tolerance, and may be combined with targeted therapies such as or for enhanced efficacy in certain cases. While primarily established for , FOLFIRI is also under investigation for other solid tumors, including gastric and pancreatic cancers. FOLFIRI's development stemmed from pivotal phase III trials in the late and early , building on the 1957 discovery of by Heidelberger and subsequent enhancements with leucovorin to boost its . A landmark 2000 multicenter randomized trial by Douillard et al. demonstrated that adding to and significantly improved median survival from 14.1 months to 17.4 months, time to progression from 4.4 to 6.7 months, and response rates from 22% to 35% compared to - alone, establishing it as a first-line standard. This infusional schedule, known as the "de Gramont" modification, reduced toxicity compared to earlier bolus regimens like IFL, making it more tolerable for long-term use. Today, FOLFIRI remains a cornerstone of metastatic therapy, often sequenced with oxaliplatin-based regimens like for optimal outcomes.

Overview

Definition and Components

FOLFIRI is a used primarily in the of metastatic , comprising the drugs (leucovorin), 5- (5-FU), and irinotecan hydrochloride. The acronym derives from "FOL" for folinic acid, "F" for 5-fluorouracil, and "IRI" for irinotecan. Leucovorin calcium, also known as , is an of folic acid and a folic acid analog that serves to potentiate the cytotoxic effects of 5-FU in the regimen. 5-Fluorouracil is a fluoropyrimidine analog and agent. Irinotecan hydrochloride is the hydrochloride salt of a semisynthetic derivative of and functions as a I inhibitor. In the standard FOLFIRI regimen, is administered at 180 mg/m² intravenously over 30-90 minutes on day 1, followed by leucovorin at 400 mg/m² intravenously over 2 hours on day 1, a 5-FU bolus of 400 mg/m² intravenously on day 1, and then 5-FU as a continuous intravenous infusion of 2400 mg/m² over 46 hours on days 1-2. The design of FOLFIRI exploits wherein leucovorin enhances 5-FU's antitumor activity, complemented by irinotecan's distinct cytotoxic mechanism to improve overall efficacy against cells.

Historical Development

The development of FOLFIRI began in the late as an effort to improve upon the IFL regimen, which combined with weekly bolus infusions of 5-fluorouracil and leucovorin but was associated with significant , particularly severe and . Researchers in , through the GERCOR group, sought to mitigate these issues by incorporating a continuous infusional schedule for 5-fluorouracil and leucovorin, drawing from established European protocols for fluoropyrimidine administration. This evolution built on the 1996 FDA accelerated approval of (under the trade name Camptosar) for second-line treatment of metastatic refractory to 5-fluorouracil-based therapy, enabling its integration into frontline combination strategies by the early 2000s. Initial French trials describing FOLFIRI emerged in 1999, with a phase II study by et al. evaluating the combination in pretreated patients, reporting promising response rates and tolerable with the infusional approach. These efforts culminated in a pivotal phase III trial published in 2000 by Douillard et al. in , which demonstrated the superiority of combined with infusional 5-fluorouracil and leucovorin (FOLFIRI) over 5-fluorouracil and leucovorin alone as first-line therapy for metastatic , establishing its efficacy with a median survival benefit. Concurrently, the U.S.-based Saltz et al. study in the Journal of Medicine that same year validated plus 5-fluorouracil and leucovorin (albeit in a weekly bolus IFL format) over 5-fluorouracil and leucovorin monotherapy, further supporting the shift toward irinotecan-containing regimens and influencing global adoption of FOLFIRI variants. The regimen's schedule evolved to incorporate the de Gramont infusional protocol for 5-fluorouracil (a bimonthly high-dose leucovorin followed by bolus and 46-48 hour continuous infusion of 5-fluorouracil), which had been optimized in earlier trials for better tolerability and efficacy compared to bolus administration. By the early 2000s, FOLFIRI gained traction in clinical protocols, with full FDA approval for in combination with 5-fluorouracil and leucovorin in April 2000 based on these studies. It became a first-line standard following the 2000 publications, particularly in , where the infusional schedule aligned with regional practices. By the mid-2000s, FOLFIRI was a recommended first-line option in guidelines, including those from ESMO, reflecting its integration into consensus treatment strategies.

Pharmacology

Mechanisms of Action

, a analog, functions as a that is metabolically activated in the liver and tumor tissues by carboxylesterases to its potent metabolite . exerts its cytotoxic effects by binding to the I-DNA cleavage complex, stabilizing it and preventing the religation of single-strand DNA breaks during replication; this leads to collisions with the replication fork, resulting in double-strand DNA breaks, S-phase arrest, and subsequent primarily in rapidly proliferating cells. 5-Fluorouracil (5-FU), a analog, interferes with synthesis through multiple mechanisms following its conversion to active metabolites. Fluorodeoxyuridine monophosphate (FdUMP), one key metabolite, forms a covalent ternary complex with (TS) and 5,10-methylenetetrahydrofolate, irreversibly inhibiting TS and depleting thymidine triphosphate (dTTP) pools essential for , thereby inducing DNA damage and cell cycle arrest. Additionally, fluorouridine triphosphate (FUTP) incorporates into , disrupting RNA processing and protein synthesis, which contributes to , particularly with bolus administration. Leucovorin (folinic acid), a reduced folate, enhances 5-FU's efficacy without direct antitumor activity by increasing intracellular levels of 5,10-methylenetetrahydrofolate, which stabilizes the FdUMP-TS inhibitory complex and prolongs TS inhibition, amplifying the disruption of DNA synthesis. This biomodulation is particularly effective in high-TS-expressing tumors, such as those in colorectal cancer, where folate supplementation shifts the equilibrium toward sustained TS blockade. The components of FOLFIRI exhibit synergistic interactions that enhance overall antitumor activity beyond additive effects. Irinotecan-induced DNA damage via I inhibition sensitizes cancer cells to 5-FU's metabolic perturbations by impairing and exacerbating nucleotide imbalances, with optimal synergy observed when irinotecan precedes 5-FU by 24 hours, allowing accumulation of replication-mediated breaks that 5-FU exacerbates through dTTP depletion. This combination is especially potent in colorectal tumors with high proliferative rates, where S-phase-specific damage from aligns with 5-FU's interference in . In specific molecular contexts, the efficacy of FOLFIRI is modulated by tumor genetic features. Mismatch repair deficiency (dMMR or MSI-H status) confers relative resistance to the regimen, particularly to the 5-FU component, due to upregulated expression and enhanced proficiency that mitigates thymineless death and mismatch errors induced by nucleotide imbalances. Conversely, mutations, common in , do not directly alter the core biochemical actions of FOLFIRI but primarily impact the efficacy of anti- targeted therapies by activating downstream survival pathways like MAPK/ERK; for FOLFIRI alone, some studies show a non-significant trend toward slightly reduced response rates and compared to RAS wild-type tumors, though the regimen retains activity independent of EGFR signaling.

Pharmacokinetics

FOLFIRI, a combination , exhibits influenced by the distinct , , , and profiles of its components—irinotecan, 5-fluorouracil (5-FU), and leucovorin—all delivered via intravenous to achieve therapeutic steady-state levels while minimizing peak concentrations. , the backbone, undergoes hepatic primarily through carboxylesterases (CES1 and CES2) to its active metabolite , which is approximately 100- to 1,000-fold more potent as a I inhibitor; is then glucuronidated by glucuronosyltransferase 1A1 (UGT1A1) to the inactive SN-38G. The elimination of ranges from 6 to 12 hours, with detectable for extended periods due to enterohepatic recirculation, and primary occurs via the biliary route, with about 66% of the dose eliminated in through ABC transporters such as ABCB1, ABCC2, and ABCG2. 5-FU demonstrates rapid , predominantly in the liver by dihydropyrimidine (DPYD), the rate-limiting that converts over 80% of the drug to inactive metabolites like dihydrofluorouracil, with subsequent breakdown to fluoro-β-alanine. Its short plasma of 8 to 20 minutes reflects this swift degradation, with clearance involving both hepatic metabolism and renal , where 7-20% is eliminated unchanged in urine, primarily within the first hour post-administration. Leucovorin, a reduced , is rapidly converted in hepatic and intestinal tissues to active forms such as 5-methyltetrahydrofolate and 5,10-methylenetetrahydrofolate, which potentiate 5-FU's effects by stabilizing the inhibitory complex; its elimination is approximately hours, with primary renal of the parent compound and metabolites. Pharmacokinetic variability in FOLFIRI is significantly impacted by genetic polymorphisms, notably UGT1A1 variants such as *28 (rs8175347), which reduce efficiency in up to 31% of individuals of ancestry, elevating irinotecan-associated toxicity risks like and . Similarly, DPYD variants, including *2A (rs3918290) and c.2846A>T (rs67376798), impair 5-FU , increasing severe toxicity odds by over fivefold in heterozygous carriers and necessitating dose adjustments. The regimen's infusion-based delivery—particularly the continuous infusion of 5-FU—helps maintain steady-state levels, reducing exposure to high peak concentrations that could exacerbate toxicity.

Clinical Applications

Indications and Contraindications

FOLFIRI is primarily indicated as a first- or second-line chemotherapy regimen for patients with metastatic colorectal cancer (mCRC), particularly those who are not candidates for oxaliplatin-based therapy or following progression on prior regimens such as FOLFOX. According to the NCCN Clinical Practice Guidelines for Colon Cancer (Version 2025), FOLFIRI, often combined with biologic agents, is recommended for initially unresectable mCRC based on tumor sidedness, RAS/BRAF mutation status, and patient performance. The ESMO Clinical Practice Guidelines for metastatic colorectal cancer, updated following key trials post-2020, endorse FOLFIRI as a doublet backbone for fit patients with Eastern Cooperative Oncology Group (ECOG) performance status of 0-2, emphasizing its role in improving progression-free survival in this setting. In select cases, FOLFIRI is also utilized for other gastrointestinal malignancies. For advanced or metastatic gastric cancer, particularly in second- or third-line settings after platinum-based therapy failure, FOLFIRI is considered per NCCN and ESMO guidelines, with evidence supporting its activity in platinum-refractory disease. Similarly, for previously treated advanced , FOLFIRI has shown modest response rates and manageable toxicity in phase II studies, though NCCN guidelines (2025) prefer regimens such as liposomal plus 5-FU/leucovorin for patients with good who have progressed on gemcitabine-based regimens. Patient selection for FOLFIRI requires careful evaluation, including molecular testing for and BRAF mutations to determine eligibility for targeted combinations, as anti-EGFR therapies are restricted to RAS wild-type tumors. Treatment is generally limited to patients with ECOG 0-2 to ensure tolerability, as per both NCCN and ESMO recommendations. Absolute s to FOLFIRI include severe dihydropyrimidine dehydrogenase (DPYD) deficiency, which predisposes to life-threatening from , necessitating either testing or avoidance per CPIC guidelines. Active is also an absolute due to the high risk of exacerbated gastrointestinal , particularly severe diarrhea from . represents another absolute , as all components (, , and ) carry risks of fetal harm, with recommendations to avoid conception for at least 6 months post-treatment. Relative contraindications include , which impairs metabolism via UGT1A1 deficiency and requires dose reduction to mitigate and risks, as outlined in ASCO pharmacogenetic guidance. Other relative factors encompass moderate hepatic or renal impairment, recent , or uncontrolled infections, where individualized risk-benefit assessment is essential per NCCN and ESMO protocols.

Regimen and Administration

The FOLFIRI regimen is administered on a standard biweekly schedule, with all components given on day 1 of a 14-day cycle. is infused intravenously at a dose of 180 /m² over 90 minutes for the first administration, with subsequent infusions potentially shortened to 30-90 minutes if tolerated; leucovorin follows concurrently at 400 /m² over 2 hours; this is immediately followed by a 5-fluorouracil (5-FU) bolus of 400 /m² intravenously, then a continuous 5-FU infusion of 2400 /m² over 46 hours. All doses are calculated based on the patient's to ensure appropriate dosing. Treatment cycles are typically continued for 6 to 12 cycles or until progression or unacceptable occurs, with the total duration often spanning up to 6 months depending on response and tolerance. Premedication includes antiemetics consistent with a moderate emetogenic risk profile, such as and dexamethasone, to prevent and vomiting. Additionally, atropine sulfate (0.25-1 mg intravenously or subcutaneously) is administered prophylactically or as needed prior to or during irinotecan infusion to counteract symptoms like abdominal cramping, diaphoresis, or early-onset . During the infusion period, patients are monitored closely for infusion-related reactions, with checked periodically; is emphasized through oral fluid intake recommendations (at least 2-3 liters daily) to prevent , particularly in anticipation of potential , and intravenous fluids may be provided if symptoms develop. Dose adjustments are made for : for 3 non-hematologic events (excluding alopecia and ), is typically reduced by 25% (to 135 mg/m²), the 5-FU bolus by 20% (to 320 mg/m²), and the 5-FU infusion by 20% (to 1920 mg/m²); further reductions or delays occur for hematologic toxicities such as . Variations to the biweekly protocol include weekly schedules with reduced doses (e.g., 80-125 mg/m², adjusted 5-FU, and leucovorin), which may be considered for patients with tolerability concerns but are less commonly used due to comparable efficacy with the standard regimen.

Combinations and Variations

Biologic Agent Combinations

FOLFIRI is commonly combined with biologic agents targeting (VEGF) or (EGFR) pathways to enhance antitumor efficacy in metastatic (mCRC). These integrations leverage complementary mechanisms: agents like inhibit tumor , reducing nutrient supply and promoting tumor that sensitizes cells to cytotoxic , while anti-EGFR monoclonal antibodies such as and block ligand binding and downstream signaling in RAS wild-type tumors, thereby augmenting and irinotecan-induced . Bevacizumab, a recombinant humanized against VEGF-A, is integrated into first-line FOLFIRI for mCRC regardless of status. This combination improves (PFS) compared to FOLFIRI alone, as demonstrated in randomized phase III trials where median PFS extended from approximately 7-8 months to 9-10 months with the addition of . The standard dosing is 5 mg/kg or 7.5 mg/kg (escalating to 10 mg/kg in some regimens) intravenously every 2 weeks, administered prior to components. The U.S. (FDA) approved in combination with fluorouracil-based regimens, including irinotecan-containing protocols like FOLFIRI, for first-line mCRC treatment in February 2004, based on pivotal data showing a 4.7-month overall survival benefit. The European Society for Medical Oncology (ESMO) guidelines endorse this regimen as a standard option for fit patients. Sequencing typically involves induction with FOLFIRI plus until disease progression or toxicity, followed by maintenance with reduced-intensity fluoropyrimidine to prolong PFS while minimizing cumulative exposure. However, recent 2023 analyses indicate no significant survival advantage from FOLFIRI plus in BRAF V600E-mutated mCRC, where outcomes remain poor with median overall survival under 12 months. As of 2025, first-line plus combined with has shown superior OS compared to standard regimens like FOLFIRI plus in BRAF V600E-mutated mCRC (HR 0.49), per phase III data. For patients with RAS wild-type mCRC, FOLFIRI is combined with anti-EGFR agents or to exploit dependency in the absence of activating RAS mutations. , a chimeric IgG1 , added to first-line FOLFIRI significantly improves overall survival (OS) in RAS wild-type tumors, with updated trial data showing a median OS of 28.4 months versus 20.2 months with FOLFIRI alone ( 0.69). Similarly, the FIRE-3 trial demonstrated an OS benefit for FOLFIRI plus over FOLFIRI plus in KRAS exon 2 wild-type mCRC (median OS 33.1 months versus 25.0 months), later confirmed in extended wild-type cohorts. yields comparable efficacy to when substituted in RAS wild-type settings, as supported by regulatory approvals and guidelines for use with FOLFIRI, though direct first-line comparative phase III data with FOLFIRI is limited. FDA approvals for with FOLFIRI in first-line KRAS wild-type mCRC occurred in 2011, expanding to full wild-type (exons 2, 3, 4 of and NRAS) by 2017 following refinements; received similar labeling updates post-2014. ESMO guidelines post-2010 emphasize testing prior to anti-EGFR initiation to predict response. Guideline recommendations, including NCCN 2025, favor anti-EGFR agents over for left-sided wild-type mCRC due to greater OS benefits observed in subgroup analyses, while is preferred for right-sided tumors. These combinations are contraindicated in RAS-mutated tumors due to lack of efficacy and potential harm.

Alternative Regimens

In metastatic colorectal cancer (mCRC), FOLFOX, comprising 5-fluorouracil (5-FU), leucovorin, and , serves as a preferred first-line regimen for patients without contraindications to , such as preexisting neuropathy. This doublet offers efficacy comparable to FOLFIRI, with similar overall survival (OS) rates around 20-24 months in combination with biologics, but it is associated with lower rates of severe compared to irinotecan-based therapy. However, FOLFOX carries a higher risk of peripheral sensory neuropathy, which can limit its duration and necessitate dose reductions or discontinuation in up to 20-30% of patients. For patients with advanced pancreatic who are fit and have good (ECOG 0-1), —a triplet regimen adding to the FOLFIRI backbone—is recommended as a first-line option, particularly for those with metastatic disease. This regimen demonstrates superior median OS (11.1 months versus 6.8 months with monotherapy) but incurs higher toxicity, including grade 3-4 (46% versus 21%), diarrhea (12% versus 1%), and fatigue (24% versus 17%), making it unsuitable for frail individuals. is thus reserved for younger, robust patients to maximize response rates while monitoring for cumulative toxicities. In elderly or frail patients with mCRC, CAPOX (capecitabine and ) or single-agent fluoropyrimidine options like monotherapy are favored to minimize toxicity while maintaining efficacy. CAPOX shows comparable OS to in adjuvant settings for those over 70 years, with tolerable neuropathy and hand-foot , though it may require dose adjustments. , while effective, is generally preferred over monotherapy in this population due to better response rates (around 30-40% versus 20-25%) and reduced risk of severe myelosuppression when combined with 5-FU. Sequential strategies, such as initiating followed by FOLFIRI upon progression, align with 2025 NCCN guidelines for optimizing outcomes in mCRC by leveraging non-cross-resistant agents. This approach yields similar median OS (approximately 23 months) regardless of sequence, allowing for prolonged exposure to both and . Key differences between FOLFIRI and alternatives center on toxicity profiles: FOLFIRI exhibits low (less than 5% grade 3) but higher gastrointestinal effects like (10-20% grade 3), contrasting with the neuropathy-dominant risks of and CAPOX (15-25% grade 3). These distinctions guide regimen selection based on patient comorbidities, with FOLFIRI often chosen to avoid cumulative nerve damage in long-term therapy.

Efficacy and Evidence

Key Clinical Trials

The pivotal phase III trial establishing the efficacy of combined with infusional 5-fluorouracil and leucovorin (FOLFIRI) in first-line treatment for metastatic (mCRC) was the multicenter, randomized study by Douillard et al., involving 430 previously untreated patients. Patients were randomized to receive either FOLFIRI or 5-FU/leucovorin alone, with treatment administered every 2 weeks until disease progression or unacceptable toxicity. Inclusion criteria encompassed histologically confirmed mCRC, measurable disease, and no prior chemotherapy for advanced disease; the primary endpoint was time to disease progression, assessed by independent review. Building on this foundation, the CRYSTAL trial was a large, international, randomized phase III study evaluating the addition of to FOLFIRI in 1,197 patients with untreated EGFR-expressing mCRC. Conducted across multiple centers, it randomized participants 1:1 to FOLFIRI plus or FOLFIRI alone every 2 weeks, with stratification by geographic region and baseline WHO performance status. Eligibility required measurable disease per RECIST criteria, no prior for metastases, and later retrospective stratification by tumor status; the primary endpoint was , with secondary endpoints including overall response rate and overall survival. The FIRE-3 trial further advanced biologic combinations with FOLFIRI through a randomized, open-label phase III design in 592 patients with KRAS 2 wild-type mCRC, comparing EGFR inhibition to VEGF inhibition. Patients, eligible if chemotherapy-naïve for advanced disease and with ECOG 0-2, were assigned 1:1 to FOLFIRI plus or FOLFIRI plus every 2 weeks until progression. Stratification factors included resectability of metastases and bevacizumab pretreatment; the primary was objective response rate by independent central review, with overall survival as a key secondary . The BREAKWATER trial is a phase III study targeting BRAF V600E-mutated mCRC, incorporating FOLFIRI in combination with targeted therapies in a multi-cohort, randomized design. Enrolling treatment-naïve patients with confirmed and measurable disease, it randomizes participants to plus with or without (including FOLFIRI or mFOLFOX6) versus standard-of-care regimens, administered every 2 weeks. Inclusion requires ECOG 0-1 and no prior for mCRC; primary endpoints are and overall survival, stratified by chemotherapy choice and co-primary for different mutation subgroups. As of 2025, results showed significant improvements with plus plus mFOLFOX6 versus standard of care: objective response rate 61% versus 40%, hazard ratio 0.53, and overall survival hazard ratio 0.49. These trials, all multicenter and randomized, typically included adults with histologically confirmed untreated mCRC, Eastern Cooperative Oncology Group performance status of 0-2, and adequate organ function, emphasizing FOLFIRI's integration into first-line strategies through rigorous endpoint evaluations like response and progression metrics.

Survival and Response Outcomes

FOLFIRI has shown objective response rates (ORR) of 30-50% in first-line treatment for metastatic (mCRC). In the seminal phase III trial by Douillard et al., FOLFIRI achieved an ORR of 35%, significantly higher than the 22% observed with 5-fluorouracil/leucovorin (5-FU/LV) alone. Similarly, the CRYSTAL trial reported an ORR of 38.8% with FOLFIRI monotherapy, which increased to 47.3% when combined with . Progression-free survival (PFS) with FOLFIRI alone typically ranges from 6.5 to 8.5 months in first-line mCRC settings. The Douillard trial established a time to progression of 6.7 months for FOLFIRI compared to 4.4 months for 5-FU/. Addition of biologic agents extends PFS to 9-11 months; for instance, in the CRYSTAL trial, plus FOLFIRI yielded a PFS of 8.9 months versus 8.0 months with FOLFIRI alone. overall survival (OS) with FOLFIRI ranges from 14 to 20 months. The Douillard trial reported a OS of 17.4 months for FOLFIRI, representing an improvement over the 14.1 months with 5-FU/ alone. In RAS wild-type patients, combination with EGFR inhibitors can achieve OS exceeding 24 months; the FIRE-3 trial demonstrated a OS of 33.1 months with plus FOLFIRI versus 25.0 months with plus FOLFIRI. A 2023 confirmed a 10-15% OS improvement with irinotecan-based regimens like FOLFIRI over 5-FU/ in first-line mCRC. Subgroup analyses indicate superior outcomes with FOLFIRI-based regimens in left-sided tumors compared to right-sided ones. In the FIRE-3 trial, patients with left-sided wild-type mCRC treated with plus FOLFIRI had a median OS of 38.7 months, versus 16.1 months for right-sided tumors. Efficacy appears limited in elderly patients over 75 years, with phase III data showing no substantial survival benefit from intensified FOLFIRI variants and higher risks in this group.

Adverse Effects

Common Side Effects

The most common side effects of FOLFIRI are gastrointestinal in nature, primarily , which occurs in two distinct forms: early-onset and delayed . Early-onset diarrhea, mediated by effects of , typically manifests within 24 hours of administration and is characterized by abdominal cramping, sweating, and loose stools; it affects up to 50% of patients but is usually mild and self-limiting. Delayed diarrhea, resulting from mucosal damage in the intestines, peaks around days 5-10 post-infusion and can be more severe, with grade 3-4 incidence ranging from 10-20% across clinical trials. In the pivotal Saltz et al. trial of combined with 5-fluorouracil and leucovorin (a regimen similar to FOLFIRI), grade 3-4 diarrhea occurred in 22.7% of patients. Risk factors for severe diarrhea include prior pelvic , which increases susceptibility to gastrointestinal toxicity. Nausea and are also frequent, affecting 50-60% of patients overall, though severe (grade 3-4) cases are less common at approximately 10%. These symptoms are often linked to both and 5-fluorouracil components and can occur shortly after . In the Saltz , grade 3-4 was reported in 9.7% of patients receiving the regimen. Hematologic toxicities include , which is severe (grade 3-4) in 20-30% of patients, with a combined incidence of 24% noted in phase III trials of FOLFIRI; this was 53.8% (grade 3: 29.8%, grade 4: 24%) in the Saltz study. occurs in about 10% of cases at grade 3 or higher, contributing to overall but generally manageable. Other common effects encompass fatigue, reported in around 40% of patients and often worsening with cumulative cycles, affecting 15-20% (primarily mild oral inflammation from 5-fluorouracil), and hand-foot syndrome in 5-10%, presenting as and tenderness on palms and soles due to infusional 5-fluorouracil. These effects are typically grade 1-2 and resolve between cycles, though monitoring is essential to prevent escalation. In the Saltz trial, grade 3-4 was 2.2%. Management of these milder effects generally involves supportive care, such as antiemetics for and for early .

Serious Risks and Management

FOLFIRI therapy carries risks of severe hematologic toxicities, including , which occurs in approximately 5% of patients and can lead to life-threatening infections. Severe , affecting up to 15% of patients at grade 3 or higher, often results in , imbalances, and potential renal complications if unmanaged. Additionally, cardiac toxicity associated with 5-fluorouracil (5-FU) manifests in 1-2% of cases, primarily as causing or myocardial ischemia, typically during or shortly after infusion. Rarer but critical risks include induced by , with an incidence below 1%, presenting as acute respiratory distress that may require in severe instances. Dihydropyrimidine dehydrogenase (DPYD) deficiency, a genetic variant affecting 3-5% of populations, predisposes patients to fatal 5-FU toxicity, including profound myelosuppression, , and neurologic effects due to impaired . Management of febrile neutropenia involves immediate administration of granulocyte colony-stimulating factor (G-CSF), such as filgrastim, alongside broad-spectrum antibiotics and hospitalization for fever exceeding 38.3°C or persistent neutropenia below 500/μL. For severe diarrhea, prompt initiation of loperamide (4 mg initially, then 2 mg after each loose stool up to 16 mg/day) is standard, with atropine used for early cholinergic symptoms like abdominal cramping; intravenous hydration and electrolyte replacement are essential to prevent dehydration. Suspected cardiac events necessitate immediate discontinuation of 5-FU, administration of nitrates or for vasospasm, and cardiac monitoring; rechallenge may be considered with uridine triacetate for DPYD-related toxicity. Dose delays or reductions follow Common Terminology Criteria for Adverse Events (NCI CTCAE) grading, such as holding therapy for grade 3 events and reducing by 20-25% for recurrent grade 4 neutropenia. Monitoring protocols include weekly complete blood counts () to detect early, with daily assessments during high-risk periods. Per 2025 National Comprehensive Cancer Network () guidelines, pre-treatment for DPYD variants (e.g., *2A, 13) and UGT1A128 is recommended to identify at-risk patients, enabling dose adjustments like 50% reduction for poor metabolizers. For diarrhea-prone patients, regular hydration status and checks (e.g., , magnesium) are advised, particularly after cycles with high exposure. Supportive care escalates to hospitalization for grade 3 or 4 toxicities, including persistent fever, requiring fluids, or respiratory compromise from lung disease. Discontinuation of FOLFIRI is indicated for persistent grade 4 events unresolved after supportive measures or confirmed DPYD deficiency with severe reactions, with transition to alternative regimens like if feasible.