Abstract
TAS-102 is a new oral anti-tumor drug, composed of a thymidine-based nucleoside analog (trifluridine: FTD) and a thymidine phosphorylase inhibitor (tipiracil hydrochloride: TPI). TAS-102 has been shown to significantly improve overall survival and progression-free survival in patients with refractory metastatic colorectal cancer (mCRC) in placebo-controlled randomized phase II and III trials. The current review summarizes mechanisms of action, pharmacokinetics/dynamics and preclinical and clinical data of TAS-102 in colorectal cancer. TAS-102 is a new salvage-line treatment option for patients with mCRC. TAS-102 is well tolerated and has great potential in future clinical drug combination therapies.
Keywords: TAS-102, metastatic colorectal cancer, chemotherapy, refractory, intolerant, phase III, salvage line
Introduction
Colorectal cancer (CRC) is the third most commonly diagnosed cancer in males and the second in females, with an estimated 1.4 million cases and 693,900 deaths occurring in the year 2012 [1]. Unfortunately, 20–30% of newly diagnosed patients present with metastatic CRC (mCRC) [2]. In addition, CRC recurs in 20–50% of patients who have undergone primary colon cancer resections with curative intent [3–5]. For most patients with mCRC, treatment is palliative and not curative, and treatment goals are to prolong overall survival (OS) and maintain quality of life as long as possible.
The past 10 years have seen unprecedented advances in mCRC treatment. For more than 50 years, 5-fluorouracil (5-FU) has been the backbone of all chemotherapy schedules, both alone and in combinations with other drugs [6–10]. The combination therapies – irinotecan with 5-FU/folinic acid (FOLFIRI) [11] or oxaliplatin with 5-FU/folinic acid (FOLFOX) [12] – have become the most effective cytotoxic regimens and resulted in significantly increased response rates and improved OS. Biological agents have further increased the effect of cytotoxic therapy, including bevacizumab [13–15], ziv-aflibercept [16], cetuximab [17,18], panitumumab [19,20] and ramucirumab [21]. We can also use regorafenib, a small-molecule inhibitor of multikinases, in salvage-line settings [22]. OS in patients with mCRC has reached a median of approximately 30 months [23–27], mainly driven by the availability of new active agents which have shown efficacy in refractory mCRC. However, there is an unmet clinical need for patients who present with disease progression after exhausting all standard therapies.
Introduction to the drug
TAS-102 is a new oral drug for patient with refractory mCRC, or who are intolerant to standard chemotherapy. TAS-102 is a combination of a thymidine-based nucleoside analog (trifluridine: FTD) and a thymidine phosphorylase (TP) inhibitor (tipiracil hydrochloride: TPI), at a molar ratio of 1 : 0.5 (weight ratio, 1 : 0.471). In this review, we summarize its new mechanism of action, pharmacokinetics and pharmacodynamics, and discuss the role of TAS-102 in future clinical practice.
Introduction to the compound
Chemistry
TAS-102 is a new oral anti-cancer drug, consisting of 2′-deoxy-5-(trifluoromethyl) uridine (FTD) as the antitumor component and 5-chloro-6-[(2-iminopyrrolidin-1-yl) methyl] pyrimidine-2,4(1H,3H)-dione monohydrochloride (TPI) which prevents the degradation of FTD at a molar ratio of 1 : 0.5 (weight ratio, 1 : 0.471). FTD has a molecular weight of 296.20 and its chemical formula is C10H11F3N2O5. TPI has a molecular weight of 279.12 and its chemical formula is C9H11ClN4O2 ·HCl. Box 1 shows the chemical structures of FTD and TPI.
Box 1. Drug summary.
| Drug name | TAS-102 | |
| Phase | Launched | |
| Indication | Colorectal cancer | |
| Pharmacology description | TAS-102 is an oral combination of a thymidine-based nucleoside analog (trifluridine) and a TP inhibitor (tipiracil hydrochloride) in a molar ratio of 1.0 : 0.5. | |
| Route of administration | Oral | |
| Chemical structure | Trifluridine | tipiracil hydrochloride | 
|  |  | |
| Pivotal trial(s) | RECOURSE study | |
Pharmacodynamics
FTD is a thymidine-based nucleoside analog with a methyl group substituted to the trifluoromethyl group at the 5′ position of thymidine (Figure 1). The thymidine analog trifluorothymidine was synthesized originally by Heidelberger et al. in 1964 [28,29], who had already shown that FTD can be phosphorylated by thymidine kinase-1 (TK1) to its active monophosphate form in uninfected tissues, to mediate cytotoxicity [30].
Figure 1. Mechanism of action of TAS-102.

Thymidine analog trifluorothymidine (FTD) is converted by thymidine kinase-1 (TK1) to its monophosphate form, F3dTMP, which is further phosphorylated to its triphosphate form, F3dTTP, which is readily incorporated into DNA. TAS-102 exerts antitumor activity primarily via FTD incorporation into DNA. Thymidine phosphorylase inhibitor (TPI) inhibits the degradation of FTD.
FTD inhibits thymidylate synthase
FTD is metabolized to monophosphorylated FTD, F3dTMP through phosphorylation by intracellular thymidine kinase. F3dTMP binds covalently to the active site of thymidylate synthase (TS) at tyrosine 146. The resulting inhibition of TS leads to intracellular depletion of dTTP and subsequent dUMP accumulation, resulting in uracil misincorporation into DNA, causing DNA damage [31]. However, F3dTMP does not form a ternary complex with the TS enzyme and 5,10-CH2-THF [32] and FTD-dependent TS inhibition rapidly declines after FTD is removed [33,34]. Therefore, this TS inhibition is not same as that of 5-FU.
FTD itself is incorporated into DNA
F3dTMP is also further phosphorylated to the triphosphate metabolite, trifluoromethyl deoxyuridine 5′-tri-phosphate (F3dTTP). F3dTTP is incorporated into DNA, primarily at sites opposite adenine [35,36]. The DNA glycosylase assay showed no detectable excision of FTD paired to adenine by any of the uracil DNA glycosylases (UDG), thymine DNA glycosylase (TDG), methyl-CpG binding domain 4 (MBD4), or fractions of HeLa cell extracts. Most FTD is incorporated at T-sites (T/A, not T/G base pairs) in cellular DNA. However, when misincorporated FTD forms an FTD:G mismatch, it becomes a target of TDG or MBD4 – eventually causing cell death because of DNA strand break formation [37]. In contrast, Matsuoka showed that FTD induced few DNA strand breaks with its massive misincorporation into DNA. FTD induced p53-dependent sustained arrest at the G2 phase, which was associated with a proteasome-dependent decrease in the Cyclin-B1 protein level and the suppression of CCNB1 and CDK1 gene expression [38]. DNA damage by FTD requires further study.
Reportedly, the inhibitory activity of FTD on DNA synthesis is about 2000-fold higher than that of dThd [39]. In addition, much more FTD was incorporated into DNA than were FdUrd and other antitumor nucleosides, Ara-C and dFdC, in a HeLa cell model [40]. Furthermore, F3dTTP incorporated into DNA was retained even at 24 h after a washing-out procedure. Although intracellular F3dTTP is rapidly eliminated from tumor cells after removal of FTD from the culture medium, almost 80% of F3dTTP, once incorporated into DNA, stays in the DNA for up to 24 h. FTD was incorporated in a time-dependent manner and not in a concentration-dependent manner [40].
The antitumor effect was examined in human tumor xenograft models. Inhibition of tumor growth continued long after the end of drug administration [41]. Furthermore, the amount of FTD incorporation in DNA and the antitumor activity of TAS-102 in xenograft models were positively and significantly correlated.
TAS-102 retains its effect on FU-related resistant cancer cells
FTD showed an anti-tumor effect against 5FU-sensitive tumor cells – and more importantly, against FU-resistant tumor cells – in a mouse xenograft model [36]. These results suggest that TAS-102 exerts its antitumor activity predominantly because of its prolonged DNA incorporation, rather than through TS inhibition. Therefore, most of its antitumor effect is thought to be induced by DNA dysfunction [33].
TPI increases bioavailability of FTD
FTD is degraded by TP in vivo, therefore the elimination half-life of FTD after intravenous administration to humans is very rapid – 12 min [42]. In monkeys, the plasma FTD level after oral administration alone was very low, suggesting extensive first-pass metabolism by the liver and intestine TP [43,44]. In addition, TP is reported to be highly expressed in human tumor tissues [45–48], therefore, inhibition of intratumoral TP is speculated to further augment the antitumor effect of FTD [43]. A variety of inhibitors have been developed for TP; TPI was found to both inhibit TP-mediated processes and to potentiate activity of various thymidine derivatives by combining with them [44]. Indeed, TPI inhibits FTD degradation in the liver and intestines after oral administration, thus improving its bioavailability [44]. The optimum ratio of FTD to TPI for oral administration was determined by measuring maximum FTD plasma levels after oral administration in mice and monkeys. The optimum ratio was 1 : 0.5 M. This ratio enabled high antitumor activity and low FTD toxicity in a mouse xenograft model [43].
TPI inhibits tumor angiogenesis
TP is also identical to platelet-derived endothelial cell growth factor (PD-ECGF) and can promote tumor growth in vivo by mechanisms that include endothelial cell migration and angiogenesis [49–52]. Angiogenesis stimulates tumor growth and metastasis of many cancers. PD-ECGF/TP is known to catabolize thymidine to thymine and 2-deoxyribose 1-phosphate (2dDR1P), which may induce secretion of oxidative stress-induced angiogenic factors, such as IL-8, VEGF and MMP-1 [53]. PD-ECGF/TP is often overexpressed in many solid tumors [48] and its levels correlate well with microvessel density in colorectal cancer [54]. PD-ECGF/TP is also a potential chemotherapeutic target in the treatment of colorectal tumors, as with VEGF and its receptors (VEGF-R). TPI may exhibit an anti-angiogenic effect mediated through inhibition of TP. High PD-ECGF/TP expression is often associated with high vessel counts in colorectal tumors that express low VEGF, in contrast to tumors with high VEGF expression and high vessel density [55]. Therefore, PD-ECGF/TP expression by infiltrating cells in human colorectal tumors provides an additional alternate mechanism for tumor neovascularization. This result suggests that TPI can improve treatment efficacy even with other anti-angiogenic therapies, such as bevacizumab.
Pharmacokinetics and metabolism
When administering TAS-102 35 mg/m2 twice daily, the mean elimination half-lives after a single dose were FTD: 1.4 h, and TPI: 2.1 h. The mean elimination half-lives at steady state were FTD: 2.0 h, and TPI: 2.4 h. FTD accumulation was 2.4-fold for area under curve (AUC) for plasma concentration over time and 1.4-fold for peak plasma concentration (Cmax) after repeated administrations. In contrast, PK parameters of TPI and an inactive form of TAS-102 did not obviously change at steady state [56]. Systemic concentrations of FTD and TPI increased linearly with higher oral doses [56]. The intestinal absorption mechanism of FTD in humans was investigated using HIEC cells as an in vitro model of human intestinal epithelial cells, in which CNT1 was found to be a major transporter responsible for intestinal absorption of FTD in humans [57]. The Cmax of FTD is decreased by 40% in fed condition compared with fasted state in humans. However, AUC of FTD was comparable between both conditions. Taking TAS-102 within 1 h after completing meals is recommended, based on the observed correlation between the increased FTD Cmax and decrease in neutrophil counts.
FTD mainly binds to human serum albumin. In vitro protein binding of FTD in human plasma is >96%, independent of drug concentration and presence of TPI. Plasma protein binding of TPI is below 8%. As with normal nucleosides, FTD can easily cross the blood–brain barrier in low micromolar concentrations and is also predominantly metabolized to TF-Thy in the brain [58].
FTD and TPI are not metabolized by cytochrome P450 enzymes. FTD is mainly eliminated by metabolism through TP to form an inactive metabolite, 5-(trifluoromethyl) uracil (FTY) [30]. No other major metabolites were detected in plasma or urine. The enzyme activities of PD-ECGF/TP, TK and TS are the most important in FTD metabolism and cytotoxic activity in cancer cells. In vivo, PD-ECGF/TP mediates FTD catabolism and TK levels are often higher in human tumor tissues than normal tissues [59], which would lead to more rapid FTD activation.
Following a single dose of TAS-102 at 60 mg, mean 48-h cumulative urinary excretion was 1.5% for unchanged trifluridine, 19.2% for FTY and 29.3% for unchanged TPI. The cumulative percentages of the FTD and TPI doses excreted in the urine from 0–10 h after drug administration were 1–8% and 19–23%, respectively [56]. A population pharmacokinetic analysis revealed that renal function and body size were the primary determinants of the PK of TAS-102; renal function should be monitored closely during treating with TAS-102; dosing of TAS-102 by body surface area is adequate to reduce the variability of exposure to FTD and TPI; the PK parameters did not significantly vary with race, age, sex or hepatic function [60].
Clinical efficacy
Early clinical study
In the year 1971, an early clinical study on FTD was carried out to evaluate its efficacy with two dosage schedules of FTD in patients with breast, lung and colon carcinomas [61]. The first regimen consisted of five daily doses ranging from 1.5 to 30 m/kg/day given by single rapid injections of FTD in 9 patients. The second regimen consisted of a dose of 2.5 mg/kg/day given for 8–13 days, given by single rapid injection, of FTD every 3 h in 32 patients. FTD produced some response in colon and breast cancer patients. A reduction of >50% in tumor size occurred in 8 of 23 patients with breast cancer and an almost complete regression occurred in 1 of 6 patients with colon cancer. However, these clinical studies were discontinued, because of the short half-life of FTD because of its rapid clearance, extensive degradation by TP and severe side effects.
FTD was also investigated as an antiviral agent and is registered as Virotic®, for use against herpes simplex virus (HSV) infections. In 1964, Kaufman et al. showed that FTD had a significant antiviral action against herpes simplex keratitis in rabbits, even in strains resistant to thymidine-based nucleoside analog (2′-deoxy-5-iododeoxyuridine) [62]. Because of its very rapid half-life, FTD was not administered systemically, but was provided as a 1% ophthalmic solution for the treatment of viral disease. FTD was approved by the Food and Drug Administration (FDA) in 1980 for the treatment of primary keratoconjunctivitis and epithelial keratitis [63].
Phase I studies (Table 1)
Table 1.
Summary of phase I studies.
| Study | Tumor type | N (CRC) | Dose (mg/m2) | Schedule | RD (mg/m2) | Dose limiting Toxicity | DCR (%) | TTF (days) | 
|---|---|---|---|---|---|---|---|---|
| 9801 | Metastatic solid tumor | 14 (14) | 50–100 | Once daily, day 1–14, every 3 weeks | 50 | Granulocytopenia | 29 | 57 | 
| 9802 | Metastatic solid tumor | 24 (20) | 50–110 | Once daily, day 1–5 and 8–12, every 4 weeks | 100 | Granulocytopenia | 29 | 63.5 | 
| 9803 | Metastatic solid tumor | 39 (32) | 100–180 | Once daily, day 1–5, every 3 weeks | 160 | Granulocytopenia | 30 | 50 | 
| 9804 | Metastatic breast cancer | 19 (0) | 50–80 | Twice daily, day 1–5 and 8–12, every 4 weeks | 50 | Granulocytopenia | 63 | 117 | 
| 9805 | Metastatic solid tumor | 15 (9) | 60–80 | Three times daily, day 1–5 and 8–12, every 4 weeks | 60 | Granulocytopenia | 60 | 123 | 
| J001 | Metastatic solid tumor | 21 (18) | 30–70 | Twice daily, day 1–5 and 8–12, every 4 weeks | 70 | Neutropenia | 52 | 92.5 | 
| Bendell et al. | Metastatic colorectal cancer | 27 (27) | 60–70 | Twice daily, day 1–5 and 8–12, every 4 weeks | 70 | Febrile neutropenia | 65 | PFS 4.1 (months) | 
CRC: colorectal cancer, RD: recommended dose, DCR: disease control rate, TTF: time to failure, PFS: progression free survival.
Phase I studies were conducted to establish the adequate dosage schedule of TAS-102 from 1998. The first phase I study (TAS102–9801) [64] treated 14 patients with advanced solid tumors once daily with TAS-102 on days 1–14 every 21 days. The recommended dose (RD) for this schedule was 50 mg/m2/day, and the dose-limiting toxicity (DLT) was granulocytopenia. In the second phase I study (TAS102–9802) [65], 24 patients with solid tumor were treated with once-daily TAS-102 on days 1–5 and 8–12 every 28 days. The RD was 100 mg/m2/day. The third phase I study (TAS102–9803) [65] evaluated once-daily TAS-102 on days 1–5 every 21 days. The DLT was granulocytopenia and RD was 160 mg/m2/day.
Other phase I studies were conducted to evaluate twice-daily or three-times-daily administration of TAS-102 because divided daily dosing of FTD resulted in higher antitumor activity in preclinical studies because of increased incorporation of FTD into DNA [40]. In the phase I study (TAS102–9804) [66], Green et al. administered to patients with metastatic breast cancer twice-daily TAS-102 on days 1–5 and 8–12 every 28 days. The DLT was granulocytopenia; the RD for heavily pretreated metastatic breast cancer was 50 mg/m2/day using this schedule. In next phase I study (TAS102–9805) [67], 15 patients with metastatic gastrointestinal cancer were treated with three times daily with TAS-102 on days 1–5 and 8–12 every 28 days. DLT was granulocytopenia and RD was 70 mg/m2/day. Based on the results derived from schedules with multiple daily doses, the phase I study (J001) was done in Japan with twice-daily TAS-102 on days 1–5 and 8–12 every 28 days [56]. The RD was 70 mg/m2/day and DLT was neutropenia. The disease control rate (DCR) was 50.0%, including 6 patients who were able to continue treatment over 12 weeks; the median progression-free survival (PFS) and OS in this cohort were 2.4 and 9.8 months, respectively. These results suggested that the efficacy of TAS-102 should be investigated using the J001 schedule in phase II trials (J003).
After the Japanese mCRC phase II trial (J003), a more recent phase I study was designed to evaluate safety and determine whether the phase II dosing and schedule was feasible in similar patients with refractory mCRC in the United States [68]. Patients were enrolled into two sequential doses (60 or 70 mg/m2/day) using a standard 3 + 3 design. No DLT was observed in the 60 mg/m2 group; one DLT (grade 3 febrile neutropenia) was observed at 70 mg/m2. Therefore, the RD was identified as 70 mg/m2/day. In conclusion, the 70 mg/m2 daily dose is generally well tolerated by patients with refractory mCRC in a Western population.
Phase II study
On the basis of the phase I data [56], TAS-102 was further evaluated in a double-blind, randomized, placebo-controlled, phase II study (J003) in 169 patients with refractory mCRC or who were intolerant of standard chemotherapy. Patients were randomly assigned in a 2 : 1 ratio to either TAS-102 (oral, 35 mg/m2, twice daily on days 1–5 and 8–12 every 28 days) or placebo [69]. All patients received best supportive care. Median OS, the primary endpoint, was 9.0 months in the TAS-102 group and 6.6 months in the placebo group (HR: 0.56; 95% CI: 0.44–0.71; p = 0.001; Table 2). Median PFS (as assessed by the independent review committee) was 2.0 months in the TAS-102 group and 1.0 month in the placebo group (HR: 0.41; 95% CI: 0.28–0.59; p < 0.0001). TAS-102 offered a significant prolongation in median OS and PFS. In the assessment by the independent review committee, 49 (43%) patients given TAS-102 achieved disease control [1 patient (1%) had a partial response and 48 patients (43%) had stable disease], as did 6 (11%) given placebo (all 6 had stable disease). The proportion of patients who achieved disease control was significantly higher in the TAS-102 group (p < 0.0001). In prespecified subgroup analyses and interaction tests for OS, PFS and DCR, the effect of TAS-102 was similar in all categories, although not all improvements were significant.
Table 2.
Phase II and III efficacy results for TAS-102.
| OS | PFS | ORR | DCR | |||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Trial | Arms | n | Time (m) | HR | 95% CI | p-value | Time (m) | HR | 95% CI | p-value | (%) | p-value | (%) | p-value | 
| J003 | Placebo | 57 | 6.6 | 0.56 | 0.44–0.71 | 0.0011 | 1.0 | 0.41 | 0.28–0.59 | <0.0001 | 0 | 1.00 | 11 | <0.0001 | 
| Phase II | TAS-102 | 113 | 9.0 | 2.0 | 1 | 43 | ||||||||
| RECOURSE | Placebo | 266 | 5.3 | 0.68 | 0.58–0.81 | <0.001 | 1.7 | 0.48 | 0.41–0.57 | <0.001 | 0.4 | 0.29 | 16 | <0.001 | 
| Phase III | TAS-102 | 534 | 7.1 | 2.0 | 1.6 | 44 | ||||||||
CI: confidence intercal; DCR: disease control rate; HR: hazard ratio; ORR: objective response rate.
In subgroup analysis, TAS-102 was likely to be more active in KRAS-mutated tumors compared with wild-type tumors; however, this subgroup analysis was limited by sample size. Therefore, the authors concluded that further investigation in future clinical studies with larger samples are warranted.
Phase III study
Based on these promising results, a multinational randomized phase III study (the RECOURSE study) was conducted to confirm the superiority of TAS-102 to placebo in the treatment of patients with refractory mCRC or who were intolerant to standard chemotherapy [70]. In this study, 800 patients were randomized to receive TAS-102 (n = 534) or placebo (n = 266) in the primary analysis. The primary endpoint, median OS, was 5.3 months in the placebo group and 7.1 months in the TAS-102 group (HR: 0.68; 95% CI: 0.58−0.81; p < 0.001; Table 2). Median PFS was 2.0 months in the TAS-102 group and 1.7 months in the placebo group (HR: 0.48; 95% CI: 0.41– 0.57; p < 0.001). These results confirmed significant OS and PFS benefits with TAS-102. Furthermore, the OS and PFS benefit with TAS-102 was consistent across all subgroups. In particular, the HRs for OS in subgroups were 0.58 in patients with KRAS wild-type tumors and 0.80 in KRAS-mutated tumors; 0.64 in Western population and 0.75 in Asian patients; 0.73 in patients with PS 0 and 0.61 in patients with PS 1; and 0.69 for patients who had already received, or not received, regorafenib – OS benefit was maintained irrespective of regorafenib use.
In multivariate Cox regression analysis, none of the factors were identified as predictive. The ORR was 1.6% (8/502) in the TAS-102 group and 0.4% (1/258) in placebo group (p = 0.29). The DCR was 44% (221/502) in the TAS-102 group and 16% (42/258) in placebo group (p < 0.001). In addition, median time to an ECOG performance status of ≥2 was 5.7 months in the TAS-102 group versus 4.0 months in the placebo group (HR: 0.66; 95% CI: 0.56–0.78; p < 0.001). TAS-102 significantly delayed worsening of disease for patients with mCRC. The percentage of patients who received post-study treatment was similar between arms (41.2% in TAS-102, 42.5% in placebo).
This phase III trial provided convincing evidence that TAS-102 is efficacious in heavily treated patients regardless of previous exposure to regorafenib and Kras status.
Safety and tolerability
Table 3 summarizes the common adverse effects of TAS-102. TAS-102 has a good safety profile. The most commonly reported adverse events of TAS-102 related to myelosuppression. During the phase I clinical trials [56,64–68], dose-limiting toxicities were grade 4 leukopenia, neutropenia and thrombocytopenia. A significant association was observed between the decrease in neutrophil count and FTD PK parameters, Cmax and AUC [56]. In the phase II study, grade 3/4 adverse events included neutropenia, leukopenia, anemia, fatigue and diarrhea. Although half of patients experienced neutropenia, the rate of febrile neutropenia was only 4%.
Table 3.
Adverse events between TAS-102 and regorafenib.
| TAS-102 | Regorafenib | |||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|
| J003 (phase II) | RECOURSE (phase III) | CORRECT | ||||||||||
| TAS-102 (n = 113) | Placebo (n = 57) | TAS-102 (n = 533) | Placebo (n = 265) | Regorafenib (n = 533) | Placebo (n = 265) | |||||||
| Adverse event, n (%) | All grade | Grade3/4 | All grade | Grade3/4 | All grade | Grade3/4 | All grade | Grade3/4 | All grade | Grade3/4 | All grade | Grade3/4 | 
| Overall adverse events | 97 | 65 | 70 | 5 | 98 | 69 | 93 | 52 | 98 | 69 | 93 | 52 | 
| Serious adverse events | 21 | 9 | 30 | 34 | 30 | 34 | ||||||
| Neutropenia | 72 | 50 | 2 | 0 | 67 | 38 | <1 | 0 | 67 | 38 | <1 | 0 | 
| Thrombocytopenia | 39 | 4 | 2 | 0 | 42 | 5 | 8 | <1 | 42 | 5 | 8 | <1 | 
| Anemia | 73 | 17 | 16 | 5 | 77 | 18 | 33 | 3 | 77 | 18 | 33 | 3 | 
| Hyperbilirubinemia | – | – | – | – | 36 | 9 | 26 | 12 | 36 | 9 | 26 | 12 | 
| Increased ALT level | – | – | – | – | 24 | 2 | 27 | 4 | 24 | 2 | 27 | 4 | 
| Increased AST level | – | – | – | – | 30 | 4 | 35 | 6 | 30 | 4 | 35 | 6 | 
| Hand-foot syndrome | – | 0 | – | – | 2 | 0 | 2 | 0 | 2 | 0 | 2 | 0 | 
| Fatigue | 58 | 6 | 42 | 4 | 35 | 4 | 23 | 6 | 35 | 4 | 23 | 6 | 
| Diarrhea | 38 | 6 | 21 | 0 | 32 | 3 | 12 | <1 | 32 | 3 | 12 | <1 | 
| Anorexia | 62 | 4 | 33 | 4 | 39 | 4 | 29 | 5 | 39 | 4 | 29 | 5 | 
| Nausea | 65 | 4 | 28 | 0 | 48 | 2 | 24 | 1 | 48 | 2 | 24 | 1 | 
| Vomiting | 34 | 4 | 25 | 0 | 28 | 2 | 14 | <1 | 28 | 2 | 14 | <1 | 
| Stomatitis | – | – | – | – | 8 | <1 | 6 | 0 | 8 | <1 | 6 | 0 | 
| Fever | – | – | – | – | 19 | 1 | 14 | <1 | 19 | 1 | 14 | <1 | 
| Asthenia | – | – | – | – | 18 | 3 | 11 | 3 | 18 | 3 | 11 | 3 | 
| Febrile neutropenia | 4 | 4 | 0 | 0 | 4 | 4 | 0 | 0 | 4 | 4 | 0 | 0 | 
ALT: alanine aminotransferase; AST: aspartate aminotransferase elevation.
A phase III study further reinforced the general tolerability of TAS-102 [70]. About 69% of patients experienced some type of grade 3/4 adverse event, which included neutropenia, leukopenia, anemia, hyperbilirubinemia and thrombocytopenia. Neutropenia was the most frequently observed clinically meaningful adverse event (grade 3/4). However, the frequency of grade 3/4 neutropenia decreased from 50% in the phase II study to 38% in the phase III study. In addition, febrile neutropenia occurred in only 4% of patients in TAS-102 group and incidence of treatment-related mortality was 0.19%. The grade 3/4 stomatitis, hand-foot syndrome, and coronary spasm, which are associated with the use of fluoropyrimidines, were seen in <1% of the patients treated with TAS-102. The adverse events were manageable and reversible. Patients were treated with symptomatic therapy and dose interruptions or reductions.
Regulatory affairs
Based on the results of the phase III clinical trial, TAS-102 was approved in Japan in March 2014, as a monotherapy to treat patients with unresectable mCRC that was refractory to standard chemotherapy. The FDA approved TAS-102 for its use in the United States in September 2015.
Conclusion
TAS-102 is an effective and well-tolerated oral anti-metabolite drug and has expanded the treatment options of patients with treatment-refractory mCRC. TAS-102 has also great potential in future clinical drug combination studies, both with cytotoxic agents (oxaliplatin and irinotecan) or targeted therapies (bevacizumab, cetuximab or panitumumab).
Expert commentary
TAS-102 has shown activity in 5-FU-resistant cells, both in in vitro and in vivo studies [36,71]. In the phase III trial, TAS-102 improved survival in patients who were refractory to 5-FU [70], which suggests that the antitumor activities of TAS-102 and 5-FU have different mechanisms [72]. DPD [73] and TS [74] are the most important biomarkers of 5-FU resistance. Increased TS levels are often associated with resistance to 5-FU [75]. FTD has another mechanism of cytotoxicity induced by TS inhibition. The antitumor activity of FTD is predominantly because of its prolonged DNA incorporation, which is an important factor in overcoming FU-resistant cancer. Although TP expression in tumors varies widely, TP expression level will not affect TAS-102 because TPI acts against colon cancer cells with both undetectable and high TP expression. In contrast, the oral 5-FU pro-drug capecitabine needs cellular TP to exert significant activity. TPI inhibits TP, which has a function in angiogenesis but also enhances the bioavailability of FTD [32,50,52]. The antitumor activity of TAS-102 in 5-FU resistant tumors might be explained by the differences in mechanism of action of FTD and 5-FU, as well as by the antiangiogenic effects of TPI.
TAS-102 and regorafenib are available for refractory mCRC and have similar efficacies. In the RECOURSE and the CORRECT [22] studies of refractory mCRC treated with standard chemotherapy, both drugs achieved more 6 month median OS time (7.1 and 6.4 months for TAS-102 and regorafenib, respectively) and DCR of 40% (44 and 41% for TAS-102 and regorafenib, respectively). Interestingly, the efficacy of TAS-102 was maintained irrespective of prior treatment with regorafenib and of Kras status. Table 2 shows the outcome of phase II/III study in TAS-102 [69,70]. The profile of adverse events differs between TAS-102 and regorafenib (Table 3). The most common adverse events are neutropenia, anemia, nausea and febrile neutropenia in patients with TAS-102, and Hand-foot syndrome, fatigue, diarrhea, hypertension, rash and liver dysfunction in patients with regorafenib. The side effect profiles of these two agents may be an important factor in the decision in what sequences these agents are used.
Five-year view
Current studies are testing combinations of TAS-102 with chemotherapy regimens such as oxaliplatin [76,77], irinotecan [78,79] docetaxel [80] bevacizumab [81] cetuximab/panitumumab [81] or erotinib [82], in CRC cell lines. Although synergic effects are seen in preclinical studies, clinical trials of combination therapy are necessary. A recent phase I/II study evaluated the efficacy of TAS-102 with bevacizumab and assessed the RD of TAS-102. It found the RD to be 70 mg/m2/day. Median PFS and OS were 5.6 months and 11.6 months, respectively, in patients with mCRC refractory to standard chemotherapy [83]. This clinical efficacy may reflect a preclinical study that showed phosphorylated FTD to increase by combining TAS-102 and bevacizumab in a CRC xenograft model [81]. Bevacizumab both inhibits angiogenesis and increases FTD accumulation and its subsequent phosphorylation in tumors. These combination therapies may be promising options.
Recent studies to identify predictive biomarker for TAS-102 showed that protein expression of two key enzymes, TK1 and TP, were not correlated with OS and PFS in patients treated with TAS-102 [84]. In contrast, Sakamoto et al. evaluated the association between the levels of FTD incorporation into DNA and the substrate specificities of hENT family members, TK1, deoxyUTPase (DUT) and DNA polymerase-α. The results indicated that FTD and FdUrd are incorporated into DNA with different efficiencies because of differences in the substrate specificities of TK1 and DUT [85]. Therefore, TK1 and DUT are potential bio-markers. Furthermore, Yamashita et al. reported that hematological toxicity may be a surrogate indicator of TAS-102 efficacy [86]. Better understanding of the TAS-102 toxicity profile and identifying the best candidates for its use, will be critical to translational development of this drug.
Key issues.
- TAS-102 is an oral anti-tumor drug, composed of a thymidine-based nucleoside analog (trifluridine: FTD) and a thymidine phosphorylase inhibitor (tipiracil hydrochloride: TPI), at a molar ratio of 1 : 0.5. 
- TAS-102 demonstrated significant improvement in OS and PFS for patients with refractory mCRC in placebo-controlled randomized phase II and III trials. 
- The most commonly reported adverse events of TAS-102 related to myelosuppression. 
- TAS-102 is US FDA-approved for treatment of colorectal cancers that had previously been treated with fluoropyrimidine, oxaliplatin, irinotecan, bevacizumab and/or EGF-receptor targeted therapy; and do not harbor KRAS mutations. 
Footnotes
Financial & competing interests disclosure
H Baba has received research funding and honoraria from Taiho Pharmaceutical Co., Ltd. HJ Lenz has received clinical support from Taiho Pharmaceutical Co., Ltd. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. Writing assistance was utilized in the production of this manuscript and performed by Edanz Group Global Ltd and funded by Y Miyamoto.
References
Papers of special note have been highlighted as:
• of interest
•• of considerable interest
- 1.Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108. [DOI] [PubMed] [Google Scholar]
- 2.Manfredi S, Lepage C, Hatem C, et al. Epidemiology and management of liver metastases from colorectal cancer. Ann Surg. 2006;244:254–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Gill S, Blackstock AW, Goldberg RM. Colorectal cancer. Mayo Clin Proc. 2007;82:114–129. [DOI] [PubMed] [Google Scholar]
- 4.Labianca R, Nordlinger B, Beretta GD, et al. Early colon cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013;24 Suppl(6): vi64–72. [DOI] [PubMed] [Google Scholar]
- 5.Watanabe T, Itabashi M, Shimada Y, et al. Japanese Society for Cancer of the Colon and Rectum (JSCCR) guidelines 2014 for treatment of colorectal cancer. Int J Clin Oncol. 2015;20:207–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Heidelberger C, Chaudhuri NK, Dannberg P, et al. Fluorinated pyrimidines, a new class of tumour-inhibitory compounds. Nature. 1957;179:663–666. [DOI] [PubMed] [Google Scholar]
- 7.Petrelli N, Douglass HO, Jr, Herrera L, et al. The modulation of fluorouracil with leucovorin in metastatic colorectal carcinoma: a prospective randomized phase III trial. gastrointestinal tumor study group. J Clin Oncol. 1989;7:1419–1426. [DOI] [PubMed] [Google Scholar]
- 8.Poon MA, O’Connell MJ, Wieand HS, et al. Biochemical modulation of fluorouracil with leucovorin: confirmatory evidence of improved therapeutic efficacy in advanced colorectal cancer. J Clin Oncol. 1991;9:1967–1972. [DOI] [PubMed] [Google Scholar]
- 9.De Gramont A, Bosset JF, Milan C, et al. Randomized trial comparing monthly low-dose leucovorin and fluorouracil bolus with bimonthly high-dose leucovorin and fluorouracil bolus plus continuous infusion for advanced colorectal cancer: a French intergroup study. J Clin Oncol. 1997;15:808–815. [DOI] [PubMed] [Google Scholar]
- 10.Kohne CH, Wils J, Lorenz M, et al. Randomized phase III study of high-dose fluorouracil given as a weekly 24-hour infusion with or without leucovorin versus bolus fluorouracil plus leucovorin in advanced colorectal cancer: European organization of research and treatment of cancer gastrointestinal group study 40952. J Clin Oncol. 2003;21:3721–3728. [DOI] [PubMed] [Google Scholar]
- 11.Saltz LB, Cox JV, Blanke C, et al. Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. Irinotecan study group. N Engl J Med. 2000;343:905–914. [DOI] [PubMed] [Google Scholar]
- 12.De Gramont A, Figer A, Seymour M, et al. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol. 2000;18:2938–2947. [DOI] [PubMed] [Google Scholar]
- 13.Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–2342. [DOI] [PubMed] [Google Scholar]
- 14.Saltz LB, Clarke S, Diaz-Rubio E, et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol. 2008;26:2013–2019. [DOI] [PubMed] [Google Scholar]
- 15.Bennouna J, Sastre J, Arnold D, et al. Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): a randomised phase 3 trial. Lancet Oncol. 2013;14:29–37. [DOI] [PubMed] [Google Scholar]
- 16.Van Cutsem E, Tabernero J, Lakomy R, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 2012;30:3499–3506. [DOI] [PubMed] [Google Scholar]
- 17.Van Cutsem E, Kohne CH, Hitre E, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009;360:1408–1417. [DOI] [PubMed] [Google Scholar]
- 18.Van Cutsem E, Lenz H-J, Kohne C-H, et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J Clin Oncol. 2015;33:692–700. [DOI] [PubMed] [Google Scholar]
- 19.Douillard J-Y, Siena S, Cassidy J, et al. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol. 2010;28:4697–4705. [DOI] [PubMed] [Google Scholar]
- 20.Douillard J-Y, Oliner KS, Siena S, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–1034. [DOI] [PubMed] [Google Scholar]
- 21.Tabernero J, Yoshino T, Cohn AL, et al. Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double-blind, multicentre, phase 3 study. Lancet Oncol. 2015;16:499–508. [DOI] [PubMed] [Google Scholar]
- 22.Grothey A, Van Cutsem E, Sobrero A, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013;381:303–312. [DOI] [PubMed] [Google Scholar]
- 23.Heinemann V, Von Weikersthal LF, Decker T, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15:1065–1075. [DOI] [PubMed] [Google Scholar]
- 24.Lenz HJ, Niedzwiecki D, Innocenti F, et al. CALGB/SWOG 80405: phase III trial of irinotecan/5-FU/leucovorin (FOLFIRI) or oxaliplatin/5-FU/leucovorin (mFOLFOX6) with bevacizumab (BV) or cetuximab (CET) for patients (pts) with expanded RAS analyses untreated metastatic adenocarcinoma of the colon or rectum (mCRC). Ann Oncol. 2014;25(Suppl. 4):A5010. [Google Scholar]
- 25.Loupakis F, Cremolini C, Masi G, et al. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal cancer. N Engl J Med. 2014;371:1609–1618. [DOI] [PubMed] [Google Scholar]
- 26.Yamada Y, Takahari D, Matsumoto H, et al. Leucovorin, fluorouracil, and oxaliplatin plus bevacizumab versus S-1 and oxaliplatin plus bevacizumab in patients with metastatic colorectal cancer (SOFT): an open-label, non-inferiority, randomised phase 3 trial. Lancet Oncol. 2013;14:1278–1286. [DOI] [PubMed] [Google Scholar]
- 27.Yamazaki K, Nagase M, Tamagawa H, et al. A randomized phase III trial of mFOLFOX6 plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment for metastatic colorectal cancer: West Japan oncology group study 4407G (WJOG4407G). J Clin Oncol. 2014;32 (5s):suppl; abstr 3534. [Google Scholar]
- 28.Heidelberger C, Parsons DG, Remy DC, Syntheses of 5-trifluoromethyluracil and 5-trifluoromethyl-2′-deoxyuridine. J Med Chem. 1964;7:1–5.• The paper describes that FTD was synthesized originally by Heidelberger et al.
- 29.Heidelberger C, King DH. Trifluorothymidine. Pharmacol Ther. 1979;6:427–442. [Google Scholar]
- 30.Heidelberger C, Boohar J, Kampschroer B. Fluorinated pyrimidines. XXIV. In vivo metabolism of 5-trifluoro-methyluracil-2-C-14 and 5-trifluoromethyl-2′-deoxyuridine-2–14. Cancer Res. 1965;25:377–381. [PubMed] [Google Scholar]
- 31.Peters GJ, Honeywell R, Leon L, et al. Role of pharmaco-dynamic, pharmacogenetic and pharmacogenomic biomarkers of cancer chemotherapy with antifolates. Pteridines. 2009;20:s115–127. [Google Scholar]
- 32.Temmink OH, Emura T, De Bruin M, et al. Therapeutic potential of the dual-targeted TAS-102 formulation in the treatment of gastrointestinal malignancies. Cancer Sci. 2007;98:779–789.• An important review devoted to TAS-102.
- 33.Tanaka N, Sakamoto K, Okabe H, et al. Repeated oral dosing of TAS-102 confers high trifluridine incorporation into DNA and sustained antitumor activity in mouse models. Oncol Rep. 2014;32:2319–2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Temmink OH, Comijn EM, Fukushima M, et al. Intracellular thymidylate synthase inhibition by trifluorothymidine in FM3A cells. Nucleos Nucleot Nucl Acids. 2004;23:1491–1494. [DOI] [PubMed] [Google Scholar]
- 35.Temmink OH, De Bruin M, Comijn EM, et al. Determinants of trifluorothymidine sensitivity and metabolism in colon and lung cancer cells. Anticancer Drugs. 2005;16:285–292. [DOI] [PubMed] [Google Scholar]
- 36.Emura T, Murakami Y, Nakagawa F, et al. A novel antimetabolite, TAS-102 retains its effect on FU-related resistant cancer cells. Int J Mol Med. 2004;13:545–549. [PubMed] [Google Scholar]
- 37.Emura T, Suzuki N, Yamaguchi M, et al. A novel combination antimetabolite, TAS-102, exhibits antitumor activity in FU-resistant human cancer cells through a mechanism involving FTD incorporation in DNA. Int J Oncol. 2004;25:571–578. [PubMed] [Google Scholar]
- 38.Matsuoka K, Iimori M, Niimi S, et al. Trifluridine induces p53-dependent sustained G2 phase arrest with its massive misincorporation into DNA and few DNA strand breaks. Mol Cancer Ther. 2015;14:1004–1013. [DOI] [PubMed] [Google Scholar]
- 39.Matulic-Adamic J, Takahashi K, Chou TC, et al. Nucleosides. 150. Synthesis and some biological properties of 5-monofluoromethyl, 5-difluoromethyl, and 5-trifluoromethyl derivatives of 2′-deoxyuridine and 2′-deoxy-2′-fluoro-beta-D-arabinofuranosyluracil. J Med Chem. 1988;31:1642–1647. [DOI] [PubMed] [Google Scholar]
- 40.Emura T, Nakagawa F, Fujioka A, et al. An optimal dosing schedule for a novel combination antimetabolite, TAS-102, based on its intracellular metabolism and its incorporation into DNA. Int J Mol Med. 2004;13:249–255. [PubMed] [Google Scholar]
- 41.Utsugi T New challenges and inspired answers for anticancer drug discovery and development. Jpn J Clin Oncol. 2013;43:945–953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Dexter DL, Wolberg WH, Ansfield FJ, et al. The clinical pharmacology of 5-trifluoromethyl-2′-deoxyuridine. Cancer Res. 1972;32:247–253. [PubMed] [Google Scholar]
- 43.Emura T, Suzuki N, Fujioka A, et al. Potentiation of the antitumor activity of alpha, alpha, alpha-trifluorothymidine by the co-administration of an inhibitor of thymidine phosphorylase at a suitable molar ratio in vivo. Int J Oncol. 2005;27:449–455.• A important study confirming the optimum ratio of FTD to TPI was 1 : 0.5 M.
- 44.Fukushima M, Suzuki N, Emura T, et al. Structure and activity of specific inhibitors of thymidine phosphorylase to potentiate the function of antitumor 2′-deoxyribonucleosides. Biochem Pharmacol. 2000;59:1227–1236. [DOI] [PubMed] [Google Scholar]
- 45.Mimori K, Matsuyama A, Yoshinaga K, et al. Localization of thymidine phosphorylase expression in colorectal carcinoma tissues by in situ RT-PCR assay. Oncology. 2002;62:327–332. [DOI] [PubMed] [Google Scholar]
- 46.El Kouni MH, El Kouni MM, Naguib FN. Differences in activities and substrate specificity of human and murine pyrimidine nucleoside phosphorylases: implications for chemotherapy with 5-fluoropyrimidines. Cancer Res. 1993;53:3687–3693. [PubMed] [Google Scholar]
- 47.Maehara Y, Nakamura H, Nakane Y, et al. Activities of various enzymes of pyrimidine nucleotide and DNA syntheses in normal and neoplastic human tissues. Gan. 1982;73:289–298. [PubMed] [Google Scholar]
- 48.Takebayashi Y, Yamada K, Miyadera K, et al. The activity and expression of thymidine phosphorylase in human solid tumours. Eur J Cancer. 1996;32A:1227–1232. [DOI] [PubMed] [Google Scholar]
- 49.Moghaddam A, Zhang HT, Fan TP, et al. Thymidine phosphorylase is angiogenic and promotes tumor growth. Proc Natl Acad Sci U S A. 1995;92:998–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hotchkiss KA, Ashton AW, Schwartz EL. Thymidine phosphorylase and 2-deoxyribose stimulate human endothelial cell migration by specific activation of the integrins alpha 5 beta 1 and alpha V beta 3. J Biol Chem. 2003;278:19272–19279. [DOI] [PubMed] [Google Scholar]
- 51.De Bruin M, Van Capel T, Van Der Born K, et al. Role of platelet-derived endothelial cell growth factor/thymidine phosphorylase in fluoropyrimidine sensitivity. Br J Cancer. 2003;88:957–964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Matsushita S, Nitanda T, Furukawa T, et al. The effect of a thymidine phosphorylase inhibitor on angiogenesis and apoptosis in tumors. Cancer Res. 1999;59:1911–1916. [PubMed] [Google Scholar]
- 53.Brown NS, Jones A, Fujiyama C, et al. Thymidine phosphorylase induces carcinoma cell oxidative stress and promotes secretion of angiogenic factors. Cancer Res. 2000;60:6298–6302. [PubMed] [Google Scholar]
- 54.Takahashi Y, Ellis LM, Mai M. The angiogenic switch of human colon cancer occurs simultaneous to initiation of invasion. Oncol Rep. 2003;10:9–13. [PubMed] [Google Scholar]
- 55.Amaya H, Tanigawa N, Lu C, et al. Association of vascular endothelial growth factor expression with tumor angiogenesis, survival and thymidine phosphorylase/platelet-derived endothelial cell growth factor expression in human colorectal cancer. Cancer Lett. 1997;119:227–235. [DOI] [PubMed] [Google Scholar]
- 56.Doi T, Ohtsu A, Yoshino T, et al. Phase I study of TAS-102 treatment in Japanese patients with advanced solid tumours. Br J Cancer. 2012;107:429–434.•A valuable phase I study demonstrating the pharmacokinetics/pharmacodynamics of TAS-102 in metastatic solid cancer patients.
- 57.Takahashi K, Yoshisue K, Chiba M, et al. Involvement of concentrative nucleoside transporter 1 in intestinal absorption of trifluridine using human small intestinal epithelial cells. J Pharm Sci. 2015;104:3146–3153. [DOI] [PubMed] [Google Scholar]
- 58.Pouremad R, Bahk KD, Shen YJ, et al. Quantitative 19F NMR study of trifluorothymidine metabolism in rat brain. NMR Biomed. 1999;12:373–380. [DOI] [PubMed] [Google Scholar]
- 59.Peters GJ, Van Groeningen CJ, Laurensse EJ, et al. A comparison of 5-fluorouracil metabolism in human colorectal cancer and colon mucosa. Cancer. 1991;68:1903–1909. [DOI] [PubMed] [Google Scholar]
- 60.Cleary JM, Mayer RJ, Cutsem E, et al. Population pharmacokinetic (PK) analysis of TAS-102 in patients (pts) with metastatic colorectal cancer (mCRC): results from 3 phase 1 trials and the phase 3 RECOURSE trial. J Clin Oncol. 2015;33s(suppl):abstr 2579. [Google Scholar]
- 61.Ansfield FJ, Ramirez G. Phase I and II studies of 2′-deoxy-5-(trifluoromethyl)-uridine (NSC-75520). Cancer Chemother Rep. 1971;55:205–208. [PubMed] [Google Scholar]
- 62.Kaufman HE, Heidelberger C. Therapeutic antiviral action of 5-trifluoromethyl-2′-deoxyridine in herpes simprex keratitis. Science. 1964;145:585–586. [DOI] [PubMed] [Google Scholar]
- 63.Carmine AA, Brogden RN, Heel RC, et al. Trifluridine: a review of its antiviral activity and therapeutic use in the topical treatment of viral eye infections. Drugs. 1982;23:329–353. [DOI] [PubMed] [Google Scholar]
- 64.Hong DS, Abbruzzese JL, Bogaard K, et al. Phase I study to determine the safety and pharmacokinetics of oral administration of TAS-102 in patients with solid tumors. Cancer. 2006;107:1383–1390.• Phase I study of TAS-102 in metastatic solid tumor with focus on mCRC.
- 65.Overman MJ, Varadhachary G, Kopetz S, et al. Phase 1 study of TAS-102 administered once daily on a 5-day-per-week schedule in patients with solid tumors. Invest New Drugs. 2008;26:445–454. [DOI] [PubMed] [Google Scholar]
- 66.Green MC, Pusztai L, Theriault LR, et al. Phase I study to determine the safety of oral administration of TAS-102 on a twice daily (BID) schedule for five days a week (wk) followed by two days rest for two wks, every (Q) four wks in patients (pts) with metastatic breast cancer (MBC). Proc Am Soc Clin Oncol. 24 18S (June 20 Supplement) 2006;24:Abstract 10576. [Google Scholar]
- 67.Overman MJ, Kopetz S, Varadhachary G, et al. Phase I clinical study of three times a day oral administration of TAS-102 in patients with solid tumors. Cancer Invest. 2008;26:794–799. [DOI] [PubMed] [Google Scholar]
- 68.Bendell JC, Rosen LS, Mayer RJ, et al. Phase 1 study of oral TAS-102 in patients with refractory metastatic colorectal cancer. Cancer Chemother Pharmacol. 2015;76:925–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yoshino T, Mizunuma N, Yamazaki K, et al. TAS-102 monotherapy for pretreated metastatic colorectal cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol. 2012;13:993–1001.• The important phase II clinical trial with reference to the use of TAS-102 in refractory mCRC confirming its efficacy in comparison to placebo.
- 70.Mayer RJ, Van Cutsem E, Falcone A, et al. Randomized trial of TAS-102 for refractory metastatic colorectal cancer. N Engl J Med. 2015;372:1909–1919.•• The most important phase III clinical trial with reference to the use of TAS-102 in refractory mCRC confirming its efficacy in comparison with placebo.
- 71.Murakami Y, Kazuno H, Emura T, et al. Different mechanisms of acquired resistance to fluorinated pyrimidines in human colorectal cancer cells. Int J Oncol. 2000;17:277–283. [DOI] [PubMed] [Google Scholar]
- 72.Lenz HJ, Stintzing S, Loupakis F. TAS-102, a novel anti-tumor agent: A review of the mechanism of action. Cancer Treat Rev. 2015;41:777–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Salonga D, Danenberg KD, Johnson M, et al. Colorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylase. Clin Cancer Res. 2000;6:1322–1327. [PubMed] [Google Scholar]
- 74.Leichman CG, Lenz HJ, Leichman L, et al. Quantitation of intratumoral thymidylate synthase expression predicts for disseminated colorectal cancer response and resistance to protracted-infusion fluorouracil and weekly leucovorin. J Clin Oncol. 1997;15:3223–3229. [DOI] [PubMed] [Google Scholar]
- 75.Peters GJ, Backus HH, Freemantle S, et al. Induction of thymidylate synthase as a 5-fluorouracil resistance mechanism. Biochim Biophys Acta. 2002;1587:194–205. [DOI] [PubMed] [Google Scholar]
- 76.Temmink OH, Hoebe EK, Van Der Born K, et al. Mechanism of trifluorothymidine potentiation of oxaliplatin-induced cytotoxicity to colorectal cancer cells. Br J Cancer. 2007;96:231–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Nukatsuka M, Nakagawa F, Takechi T. Efficacy of combination chemotherapy using a novel oral chemotherapeutic agent, TAS-102, with oxaliplatin on human colorectal and gastric cancer xenografts. Anticancer Res. 2015;35:4605–4615. [PubMed] [Google Scholar]
- 78.Temmink OH, Hoebe EK, Fukushima M, et al. Irinotecan-induced cytotoxicity to colon cancer cells in vitro is stimulated by pre-incubation with trifluorothymidine. Eur J Cancer. 2007;43:175–183. [DOI] [PubMed] [Google Scholar]
- 79.Nukatsuka M, Nakagawa F, Saito H, et al. Efficacy of combination chemotherapy using a novel oral chemotherapeutic agent, TAS-102, with irinotecan hydrochloride on human colorectal and gastric cancer xenografts. Anticancer Res. 2015;35:1437–1445. [PubMed] [Google Scholar]
- 80.Bijnsdorp IV, Kruyt FA, Gokoel S, et al. Synergistic interaction between trifluorothymidine and docetaxel is sequence dependent. Cancer Sci. 2008;99:2302–2308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Tsukihara H, Nakagawa F, Sakamoto K, et al. Efficacy of combination chemotherapy using a novel oral chemotherapeutic agent, TAS-102, together with bevacizumab, cetuximab, or panitumumab on human colorectal cancer xenografts. Oncol Rep. 2015;33:2135–2142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bijnsdorp IV, Kruyt FA, Fukushima M, et al. Molecular mechanism underlying the synergistic interaction between trifluorothymidine and the epidermal growth factor receptor inhibitor erlotinib in human colorectal cancer cell lines. Cancer Sci. 2010;101: 440–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kuboki Y, Nishina T, Shinozaki E, et al. An investigator initiated multicenter phase I/II study of TAS-102 with bevacizumab for metastatic colorectal cancer refractory to standard therapies (C-TASK FORCE). J Clin Oncol. 2015;33(suppl):abstr 3544. [DOI] [PubMed] [Google Scholar]
- 84.Komatsu Y, Yoshino T, Mizunuma N, et al. The value of thymidine kinase 1 (TK1) and thymidine phosphorylase (TP) expression as predictive factors with the treatment efficacy of TAS-102, a novel antitumor agent, in patients (pts) with metastatic colorectal cancer (mCRC). Eur J Cancer. 2011;47(suppl. 1):s421. [Google Scholar]
- 85.Sakamoto K, Yokogawa T, Ueno H, et al. Crucial roles of thymidine kinase 1 and deoxyUTPase in incorporating the antineoplastic nucleosides trifluridine and 2′-deoxy-5-fluorouridine into DNA. Int J Oncol. 2015;46:2327–2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Yamashita F, Komoto I, Oka H, et al. Exposure-dependent incorporation of trifluridine into DNA of tumors and white blood cells in tumor-bearing mouse. Cancer Chemother Pharmacol. 2015;76:325–333. [DOI] [PubMed] [Google Scholar]
