Skip Navigation
Skip to contents

Cancer Res Treat : Cancer Research and Treatment

OPEN ACCESS

Articles

Page Path
HOME > Cancer Res Treat > Volume 57(1); 2025 > Article
Original Article
Breast cancer
Endoxifen Concentration Is Associated with Recurrence-Free Survival in Hormone-Sensitive Breast Cancer Patients
Beomki Lee1,a)orcid, Seok Jin Nam2, Seok Won Kim2, Jonghan Yu2, Byung-Joo Chae2, Se Kyung Lee2, Jai Min Ryu2, Jeong Eon Lee2,orcid, Soo-Youn Lee1,orcid
Cancer Research and Treatment : Official Journal of Korean Cancer Association 2025;57(1):140-149.
DOI: https://doi.org/10.4143/crt.2023.1285
Published online: June 18, 2024

1Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea

2Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea

Correspondence: Soo-Youn Lee, Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
Tel: 82-2-3410-1834 Fax: 82-2-3410-2719 E-mail: suddenbz@skku.edu
Co-correspondence: Jeong Eon Lee, Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
Tel: 82-2-3410-0260 Fax: 82-2-3410-6982 E-mail: jeongeon.lee@samsung.com
a)Present address: Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
• Received: December 5, 2023   • Accepted: June 16, 2024

Copyright © 2025 by the Korean Cancer Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 960 Views
  • 89 Download
prev next
  • Purpose
    The metabolism of tamoxifen is influenced by various cytochrome p450 enzymes, including CYP2D6 and CYP2C19, leading to variations in the levels of endoxifen, even with the same tamoxifen dose. However, the clinical significance of endoxifen for the prognosis of breast cancer patients remains controversial. This study aimed to elucidate the relevance of endoxifen level to recurrence-free survival censored with tamoxifen discontinuation (RFSt), representing the RFS for tamoxifen itself, of breast cancer patients and determine a suitable cutoff for prognostication.
  • Materials and Methods
    The study included 478 breast cancer patients. Tamoxifen and its metabolites, including endoxifen, were measured using liquid chromatography-tandem mass spectrometry. An optimal cutoff was determined with maximally selected rank statistics. Survival analysis and Cox regression were conducted based on this cutoff.
  • Results
    An endoxifen level of 21.00 ng/mL was the optimal cutoff for prognostication. Survival analysis revealed a statistically significant difference in RFSt between the low endoxifen group (≤ 21.00 ng/mL) and the high endoxifen group (> 21.00 ng/mL) (log-rank test, p=0.032). The 10-year probability of RFSt was 83.2% (95% confidence interval [CI], 77.0 to 89.9) and 88.3% (95% CI, 83.3 to 93.5) in the low and high endoxifen groups, respectively. Multivariable Cox proportional hazards regression indicated endoxifen concentration as a significant factor associated with prognosis.
  • Conclusion
    Endoxifen could serve as a marker for appropriate tamoxifen treatment with a cutoff of 21.00 ng/mL. Based on this cutoff, therapeutic drug monitoring would benefit patients displaying suboptimal endoxifen concentrations.
Endoxifen is the most important active metabolite of tamoxifen, a drug used for treatment of hormone-sensitive breast cancer patients. While estrogen stimulates proliferation of estrogen receptor (ER)–positive breast cancer cells, tamoxifen and its metabolites render an antagonistic effect to suppress cancer cell proliferation by inhibiting expression of various genes [1]. However, even with the suppressive effect of tamoxifen on hormone-sensitive breast cancer, one-third of breast cancer patients treated with tamoxifen will experience relapse [2].
Cytochrome P450 (CYP) 2D6 is the major enzyme involved in the metabolism of tamoxifen. There have been multiple studies with conflicting results regarding the prognostic value of CYP2D6 genotype [3,4]. Although early studies demonstrated a positive association between CYP2D6 genotype and clinical outcome [5,6], subsequent studies reached a consensus that CYP2D6 genotype poorly correlates with the prognosis of breast cancer patients [7-9]. Hence, attempts were made to predict the clinical outcome based on endoxifen concentration rather than CYP2D6 genotype. Therapeutic drug monitoring (TDM) of endoxifen concentration could be beneficial for breast cancer patients if endoxifen concentration correlates with prognosis. However, there is no consensus on the cutoff for endoxifen to distinguish prognosis, with multiple studies proposing different cutoff values [10-14]. While a previous study also attempted to elucidate the prognostic significance of endoxifen using various previously reported cutoffs ranging from 3.3 to 10.3 ng/mL, none of these cutoffs were significant in the survival analysis [14].
One of the reasons for these conflicting results could be largely varying endoxifen concentrations across different populations of various studies. In a previous study incorporating a subset of the present cohort [15], the mean endoxifen concentration of patients treated with 20 mg/day tamoxifen was 25.00 ng/mL (66.93 nM). However, regarding the mean endoxifen concentration of patients treated with 20 mg/day, studies conducted in the Netherlands (the TOTAM study) [16] and Australia (the TADE study) [17], which mostly involved Caucasians, reported concentrations of 12.06 ng/mL (32.30 nM) and 9.63 ng/mL (25.80 nM), respectively. Meanwhile, a previous study, comprising Vietnamese and Filipino patients treated with 20 mg/day tamoxifen, reported a mean endoxifen concentration of 35.40 ng/mL (94.78 nM) [18]. The main reason for higher endoxifen concentrations and cutoff in our study population could be the different distributions of metabolic activity in the Korean population compared to other ethnicities. It has been reported that the frequency of poor metabolizers is significantly lower in the Korean population compared to Caucasians and Africans [19]. In addition, it has been suggested that using a relatively non-selective liquid chromatography-tandem mass spectrometry (LC-MS/MS) method could result in an endoxifen level twice as higher than that measured with a highly selective LC-MS/MS method [20].
Regardless of what contributes to the distribution of endoxifen concentration, it is likely that an appropriate endoxifen cutoff should be determined for each specific population. Considering the high endoxifen concentration in our previous study [15], we hypothesized that a higher cutoff might lead to superior predictability regarding the prognosis of hormone-sensitive breast cancer patients receiving tamoxifen treatment. In this study, we explored an endoxifen cutoff for prognostication and evaluated this presumed cutoff along with other cutoffs suggested in previous studies.
1. Study population and procedure
This study was a single-center observational study conducted at Samsung Medical Center, Seoul, Korea. The patients were enrolled from December 2009 to January 2016. The study population consisted of female patients diagnosed with breast cancer and receiving adjuvant tamoxifen treatment, which arrived at a total of 573 patients fully encompassing the 281 patients from our previous study [15]. Of note, the initial adjuvant endocrine therapy for all patients started with tamoxifen, not an aromatase inhibitor. Refer to our previous publication for detailed inclusion and exclusion criteria for the study population [15]. In this study, patients diagnosed with carcinoma in situ (with or without microinvasion) or those with poor compliance were additionally excluded. Poor compliance was determined based on patient self-report of low adherence or an endoxifen level lower than the limit of detection. In case poor compliance was identified, the information on the date of discontinuation or irregular administration of tamoxifen was also collected. In case of follow-up loss and tamoxifen treatment interruption due to various reasons, the corresponding cases were right censored. Additionally, those who received an escalated tamoxifen dose were right censored at the time of dose escalation. Clinicopathological information of age at diagnosis, pathologic findings, bilaterality, ER and progesterone receptor (PR) status, ERBB2 status, pathologic stage, tamoxifen dose, and other treatment modalities was collected. Menopausal status was not collected since we could not confidently determine the corresponding information from the medical records. The pathologic stage of each patient was classified according to the 7th edition of the American Joint Committee on Cancer (AJCC) staging manual. In patients with synchronous bilateral breast cancer, the higher pathologic stage of either side was used. ER and PR were considered positive if at least 1% of tumor cells were positive for nuclear staining. The ERBB2 status was considered positive for grade 3+ in immunohistochemical (IHC) staining or grade 2+ in IHC staining with positive in situ hybridization, but was otherwise considered negative. The in vivo endoxifen concentration was considered to be at a steady-state level if venous sampling was conducted at least eight weeks after the initiation of tamoxifen treatment. Extended hormone therapy was defined as having been treated with tamoxifen or aromatase inhibitor after finishing 5 years of tamoxifen administration. Concentrations of tamoxifen and its metabolites, which include (Z)-endoxifen, N-desmethyltamoxifen, and (Z)-4-hydroxytamoxifen, were measured from plasma with LC-MS/MS using an API 4000 tandem mass spectrometer (AB Sciex, Foster City, CA) in tandem with an Agilent Technologies Series 1200 HPLC system (Agilent Technologies, Santa Clara, CA). Details regarding the measurement procedure are described in our previous publication [15].
2. Statistical analysis
The primary endpoint in this study was recurrence-free survival (RFS) since the purpose of using tamoxifen is to suppress cancer cell proliferation and prevent relapse. To determine an optimal cutoff for endoxifen concentration in predicting RFS, maximally selected rank statistics was utilized. Using the presumed optimal cutoff, patients were divided into low endoxifen and high endoxifen groups. In reference to a previous study, RFS censored following tamoxifen discontinuation (RFSt) and complete RFS (RFSc) were evaluated [9]. RFSt represents the RFS for tamoxifen per se, and all patients with tamoxifen treatment interruption were right censored at the corresponding time point for RFSt regardless of the reason [9]. In addition, if tamoxifen dose was increased, these patients were also right censored at the time point of dose escalation. On the other hand, if the patients changed the tamoxifen dose or switched to an aromatase inhibitor, these patients were not censored for RFSc and the full period of hormone therapy was taken into account [9]. Notably, the period of observation after tamoxifen discontinuation could potentially confound the results. Since this study focused on the association between endoxifen concentration and prognosis, RFSt was mainly evaluated [9]. Regarding the baseline characteristics of the two groups, comparisons were conducted with t test and chi-square test as appropriate. Survival analysis was conducted with the Kaplan-Meier estimator, and the statistical significance of the difference between the two groups was determined with a log-rank test. Along with the presumed cutoff, endoxifen cutoffs suggested in previous studies [10-14] were evaluated with our cohort. The evaluated cutoffs from the literature were as follows: (1) 3.36 ng/mL (9.00 nM) [10], (2) 5.29 ng/mL (14.15 nM) [11], (3) 5.60 ng/mL (15.00 nM) [12], (4) 5.97 ng/mL (15.98 nM) [13], and (5) 10.30 ng/mL (27.58 nM) [14].
Cox proportional hazards regression was used to investigate other clinicopathological factors associated with increased risk of breast cancer relapse. Backward elimination was used to select the variables and build the model. The proportional hazards assumption was verified using scaled Schoenfeld residuals.
All statistical analyses were carried out with R ver. 4.3.0 (R Foundation for Statistical Computing, Vienna, Austria) with a two-sided significance level of p=0.05; plots were illustrated with ggplot2 3.4.2, maximally selected rank statistics was implemented with maxstat 0.7-25, and survival analysis and Cox proportional hazards regression analysis were performed with survival 3.5-5 and survminer 0.4.9 in R 4.3.0 (R Foundation for Statistical Computing).
1. Demographics
From December 2009 to January 2016, 573 breast cancer patients were enrolled at Samsung Medical Center, Korea. Among the 573 patients, 95 subjects were excluded, and 478 patients were analyzed in this study (Fig. 1). The 478 eligible subjects had a median follow-up period of 98.5 months (interquartile range [IQR], 57.0 to 120.8 months) with respect to RFSt. The median duration from tamoxifen treatment initiation to sample collection was 5.0 months (IQR, 4.5 to 6.0 months). The subjects were divided into two groups based on an endoxifen cutoff of 21.00 ng/mL (56.22 nM), the determining method of which will be described in the following section. The clinical characteristics of the subjects are listed in Table 1, and there were no statistically significant differences between any characteristics of the two groups except for luteinizing hormone-releasing hormone (LHRH) agonist treatment (χ2 test, p=0.029). There were no patients who underwent bilateral oophorectomy during the observed period.
2. Prognostic significance of endoxifen concentration
A presumed endoxifen cutoff of 21.00 ng/mL (56.22 nM) was defined with maximally selected rank statistics, which exhibited the highest standardized log-rank statistic value. The histogram based on this cutoff (21.00 ng/mL) is presented in Fig. 2A. However, as the threshold determined with the highest log-rank statistics does not necessarily guarantee a statistically significant difference in the primary endpoint, RFSt in our study, further investigation of this presumed cutoff was carried out with survival analysis.
Of the 478 subjects, 56 (11.7%) experienced relapse before October 2022. In the low endoxifen group (≤ 21.00 ng/mL), 29 of 149 patients (19.5%) experienced relapse, compared to 27 of 329 patients (8.2%) in the high endoxifen group (> 21.00 ng/mL). An endoxifen cutoff of 21.00 ng/mL was able to distinguish the prognosis represented by RFSt with statistical significance (log-rank test, p=0.032) (Fig. 2B). The 10-year probability of RFSt was 83.2% (95% confidence interval [CI], 77.0 to 89.9) and 88.3% (95% CI, 83.3 to 93.5) in the low and high endoxifen groups, respectively. On the other hand, when RFSc was evaluated with the same endoxifen cutoff, the difference was not statistically significant (log-rank test, p=0.127) (Fig. 2C). The 10-year probability of RFSc was 86.2% (95% CI, 81.1 to 91.6) and 89.2% (95% CI, 84.8 to 93.8) in the low and high endoxifen groups, respectively.
When the cutoffs for endoxifen suggested in previous literature were applied to our study population, relatively few patients were assigned to the low endoxifen group, which limited the evaluation of these cutoffs. As applying the cutoff of 5.97 ng/mL already resulted in fewer than five subjects being assigned to the low endoxifen group, cutoffs lower than 5.97 ng/mL were not further evaluated. For the survival analysis curve illustrated using cutoffs of 5.97 ng/mL and 10.30 ng/mL, refer to Fig. 3. There was a statistically significant difference in RFSt according to the cutoff of 5.97 ng/mL (log-rank test, p=0.047) (Fig. 3A), but not in RFSc (log-rank test, p=0.280) (Fig. 3B). However, the cutoff of 10.30 ng/mL failed to exhibit statistically significant differences in both RFSt (log-rank test, p=0.632) (Fig. 3C) and RFSc (log-rank test, p=0.516) (Fig. 3D).
3. Cox proportional hazards regression
Using backward elimination, the variables selected in the final model were endoxifen, ER status, PR status, pathologic stage, adjuvant trastuzumab treatment, LHRH agonist treatment, and extended hormone therapy. Results of the univariable and multivariable Cox proportional hazards regression model are listed in Table 2. In the univariable analysis, a significantly different hazard ratio (HR) of RFSt was associated with low endoxifen level (≤ 21.00 ng/mL; HR, 1.78; 95% CI, 1.04 to 3.04; p=0.034). In addition, ER status, PR status, and adjuvant trastuzumab treatment were significantly associated with RFSt in the univariable analysis. In the multivariable analysis, endoxifen concentration was still a significant factor associated with RFSt (HR, 1.78; 95% CI, 1.03 to 3.08; p=0.038). Besides, ER status (HR, 4.53; 95% CI, 1.07 to 19.26; p=0.041), PR status (HR, 4.92; 95% CI, 1.87 to 12.93; p=0.001), adjuvant trastuzumab treatment (HR, 3.00; 95% CI, 1.26 to 7.15; p=0.013), LHRH agonist treatment (HR, 2.45; 95% CI, 1.23 to 4.86; p=0.011), and extended hormone therapy (HR, 0.45; 95% CI, 0.23 to 0.85; p=0.015) were significantly associated with RFSt in the multivariable analysis.
CYP2D6 is the key enzyme of tamoxifen metabolism, and a new CYP2D6 locus was recently discovered as the principal determinant of endoxifen concentrations [21]. Still, enzyme activity determined with CYP2D6 genotyping has limited value in estimating the efficacy of tamoxifen since tamoxifen metabolism is affected by various factors such as age, weight, concurrently used drugs, and activity of other enzymes including CYP2C9, CYP2C19, CYP3A4, CYP3A5, and CYP2B6 [21-23], complicating the prediction of endoxifen concentration. Currently, the current National Comprehensive Cancer Network (NCCN) guideline does not recommend CYP2D6 genotyping in breast cancer patients [24]. While the Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline suggests dose escalation to 40 mg/day in CYP2D6 intermediate metabolizers and poor metabolizers [25], it was insufficient to achieve the endoxifen concentration observed in normal metabolizers [26]. Considering this situation, the current CYP2D6-based dose adjustment has limited value, and endoxifen-based dose adjustment seems more plausible. However, regarding the clinical significance of endoxifen concentration, there is no consensus on the prognostic value of endoxifen concentration [26], with various studies suggesting different endoxifen cutoff values [10-14]. Furthermore, varying endoxifen distribution in different populations [15-19] as well as discrepant endoxifen levels depending on the method of measurement [20] warrant a specific endoxifen cutoff for different contexts.
Even though tamoxifen is widely used for breast cancer patients, research on its efficacy is challenging owing to reasons such as (1) plans for pregnancy, (2) side effects such as depression and liver enzyme elevation, (3) poor compliance and failure to complete the 5-year course, (4) switch to an aromatase inhibitor, (5) extended tamoxifen treatment for 10 years, and (6) extended aromatase inhibitor treatment after finishing 5-year tamoxifen treatment. While concurrent drugs such as selective serotonin reuptake inhibitors are known to affect the metabolism of tamoxifen, the results of our study would not be affected by such drugs as we measured the concentration of endoxifen itself, the most potent active metabolite of the original drug.
Recently, the feasibility of increasing endoxifen concentration through TDM has been proposed [16,27]. To our knowledge, this study is the first to report the clinical relevance of endoxifen concentration and prognosis of breast cancer in the Korean population. Our study recommends an endoxifen concentration greater than 21.00 ng/mL considering its association with RFSt, which is a novel cutoff higher than those described in previous reports. Accomplishing this goal would be plausible using TDM as higher tamoxifen dose results in elevated endoxifen concentration [17]. Both inter-individual differences in endoxifen concentration and varying characteristics depending on the duration of hormone therapy substantiate the need for a tailored approach to tamoxifen dose adjustment.
While LHRH agonist treatment status differed significantly between the low and high endoxifen groups, endoxifen concentration remained a significant predictor in multivariable Cox regression adjusted for other clinically important variables. Regarding the various treatment modalities, low endoxifen concentration, adjuvant trastuzumab treatment, and LHRH agonist treatment were associated with higher HR, whereas hormone therapy extension was associated with lower HR in the multivariable Cox regression model for RFSt. We suspect that the higher risk in trastuzumab-treated patients would be associated with the relatively aggressive nature of ERBB2-positive breast cancer compared to ERBB2-negative breast cancer [28]. Besides, we acknowledge that the higher risk associated with LHRH agonist treatment is rather counterintuitive. Still, we suspect that varying age distribution and adjuvant chemotherapy treatment may have contributed to the poor prognosis observed in patients receiving LHRH agonist treatment. In our study population, patients treated with LHRH agonist had lower age (median, 41.2; IQR, 39.2 to 44.2) compared to those who did not receive LHRH agonist treatment (median, 44.2; IQR, 40.4 to 47.0; Wilcoxon test, p < 0.001). Thus, the poor prognosis in patients on LHRH agonist treatment could be due to the relatively poor prognosis in young breast cancer [29,30]. Moreover, although pathologic stage did not differ significantly between patients treated with and without LHRH agonist, a higher proportion of patients treated with LHRH agonist were on adjuvant chemotherapy (65.3%) compared to those who were not (38.8%; χ2 test, p < 0.001). This implies that varying clinical characteristics between the two groups, which would have led to different treatment choices, as well as the administration of adjuvant chemotherapy itself may also have contributed to the poor prognosis in patients treated with LHRH agonist. We would not address the prognostic effect of variables other than endoxifen concentration in further detail as it falls beyond the scope of our study.
In this study, we established a novel cutoff for endoxifen (21.00 ng/mL). As we had suspected, previously suggested cutoffs were unable to distinguish prognosis in our study population. Nevertheless, adopting the novel cutoff of our study in a different setting should be done with caution. We necessitated the need for a higher endoxifen cutoff because endoxifen concentration could vary across different populations [15-19] and measurement methods [20]. This logic also applies to other potential groups that plan to utilize our study in predicting the prognosis of breast cancer patients. Here, we would like to emphasize the significance of establishing a population-specific and institute-specific cutoff for endoxifen. Further validation would be required to determine the generalizability of our new endoxifen cutoff, as our study is the first to validate the prognostic significance of such a high endoxifen cutoff.
There are some limitations in this study to declare. First, the number of samples drawn from each patient was limited. Due to this limitation, there might be some noise in our data. For instance, if a patient adds a new medication affecting tamoxifen metabolism after measuring endoxifen concentration, this change would not be reflected in the data. Although we had no means to identify prescriptions from other hospitals, a meticulous review of our medical records was conducted to exclude medications from our institute affecting the CYP2D6 metabolism. Second, while menopausal status is an important factor that affects the prognosis of breast cancer patients, our study did not account for menopausal status since we couldn’t confidently obtain the relevant information from the medical records. Third, only a small number of patients received tamoxifen monotherapy. While other treatment modalities could potentially confound the effect of tamoxifen treatment, we addressed this through multivariable Cox proportional hazards regression, which showed that endoxifen concentration is still a significant factor associated with RFSt even with other clinicopathological factors adjusted.
In summary, an endoxifen cutoff (21.00 ng/mL) higher than previously suggested thresholds could be utilized in prognostication of breast cancer patients. Individualized tamoxifen dose adjustment would be beneficial for breast cancer patients to improve their prognosis by elevating the endoxifen concentration.

Ethical Statement

This study was approved by the Institutional Review Board of Samsung Medical Center (IRB No. 2011-04-030-001). Informed consent was obtained from each participant.

Author Contributions

Conceived and designed the analysis: Lee B, Lee JE, Lee SY.

Collected the data: Lee JE, Lee SY.

Contributed data or analysis tools: Nam SJ, Kim SW, Yu J, Chae BJ, Lee SK, Ryu JM, Lee JE, Lee SY.

Performed the analysis: Lee B, Lee SY.

Wrote the paper: Lee B, Lee SY.

Conflicts of Interest

Conflict of interest relevant to this article was not reported.

Acknowledgements
This research was supported by a grant of the Korea Health Technology R&D Project administered through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI14C3418). The funder had no role in the study design, analysis, and manuscript preparation and publication.
Fig. 1.
Study design.
crt-2023-1285f1.jpg
Fig. 2.
(A) Distribution of endoxifen concentration in our study population. Kaplan-Meier curve of RFSt (B) and RFSc (C) probability of breast cancer patients according to an endoxifen cutoff of 21.00 ng/mL. RFSc, complete recurrence-free survival; RFSt, recurrence-free survival censored following tamoxifen discontinuation.
crt-2023-1285f2.jpg
Fig. 3.
Kaplan-Meier curve of RFSt (A, C) and RFSc (B, D) probability of breast cancer patients according to an endoxifen cutoff of 5.97 ng/mL (15.98 nM) (A, B) and 10.30 ng/mL (27.58 nM) (C, D). RFSc, complete recurrence-free survival; RFSt, recurrence-free survival censored following tamoxifen discontinuation.
crt-2023-1285f3.jpg
Table 1.
Clinical characteristics of the study population
Characteristic Total (n=478) Low endoxifen (n=218) High endoxifen (n=260) p-valuea)
Age at diagnosis (yr) 43.8 (40.2-46.8) 43.4 (40.0-46.5) 44.0 (40.4-47.0) 0.280
Histologic finding
 IDC 397 (83.1) 182 (83.5) 215 (82.7) 0.298
 ILC 19 (4.0) 12 (5.5) 7 (2.7)
 Mixed 28 (5.9) 10 (4.6) 18 (6.9)
 Others 34 (7.1) 14 (6.4) 20 (7.7)
ER status
 ER+ 473 (99.0) 215 (98.6) 258 (99.2) 0.661
 ER– 5 (1.0) 3 (1.4) 2 (0.8)
PR status
 PR+ 464 (97.1) 209 (95.9) 255 (98.1) 0.175
 PR– 14 (2.9) 9 (4.1) 5 (1.9)
ERBB2 status
ERBB2+ 38 (7.9) 17 (7.8) 21 (8.1) > 0.99
ERBB2 440 (92.1) 201 (92.2) 239 (91.9)
Pathologic stage
 Stage I 233 (48.7) 96 (44.0) 137 (52.7) 0.112
 Stage II 181 (37.9) 87 (39.9) 94 (36.2)
 Stage III 64 (13.4) 35 (16.1) 29 (11.2)
Bilaterality
 UBC 460 (96.2) 213 (97.7) 247 (95.0) 0.153
 SBBC 18 (3.8) 5 (2.3) 13 (5.0)
Neoadjuvant chemotherapy
 Not done 473 (99.0) 217 (99.5) 256 (98.5) 0.382
 Done 5 (1.0) 1 (0.5) 4 (1.5)
Adjuvant chemotherapy
 Not done 187 (39.1) 80 (36.7) 107 (41.2) 0.347
 Done 291 (60.9) 138 (63.3) 153 (58.8)
Adjuvant trastuzumab treatment
 Not done 455 (95.2) 206 (94.5) 249 (95.8) 0.531
 Done 23 (4.8) 12 (5.5) 11 (4.2)
Adjuvant radiotherapy
 Not done 105 (22.0) 53 (24.3) 52 (20.0) 0.267
 Done 373 (78.0) 165 (75.7) 208 (80.0)
LHRH agonist treatment
 Not done 398 (83.3) 191 (87.6) 207 (79.6) 0.029
 Done 80 (16.7) 27 (12.4) 53 (20.4)
Extended hormone therapy
 Not done 344 (72.0) 157 (72.0) 187 (71.9) > 0.99
 Done 134 (28.0) 61 (28.0) 73 (28.1)

Values are presented as median (IQR) or number (%). ER, estrogen receptor; IDC, invasive ductal carcinoma; ILC, invasive lobular carcinoma; IQR, interquartile range; LHRH, luteinizing hormone-releasing hormone; PR, progesterone receptor; SBBC, synchronous bilateral breast cancer; UBC, unilateral breast cancer.

a) p-values were calculated with t tests and χ2 tests for numerical and categorical variables, respectively.

Table 2.
Univariable and multivariable Cox proportional hazards regression analyses for RFSt
Characteristic Univariable analysis
Multivariable analysis
HR (95% CI) p-value HR (95% CI) p-value
Endoxifen
 Low (≤ 21.00 ng/mL) 1.78 (1.04-3.04) 0.034 1.78 (1.03-3.08) 0.038
 High (> 21.00 ng/mL) 1 (Reference) 1 (Reference)
Age
 Per year 0.97 (0.92-1.01) 0.165
Histologic finding
 IDC 1 (Reference)
 ILC 2.29 (0.82-6.37) 0.113
 Mixed 0.33 (0.05-2.41) 0.276
 Others 1.40 (0.55-3.53) 0.476
ER status
 ER+ 1 (Reference) 1 (Reference)
 ER– 4.53 (1.10-18.65) 0.036 4.53 (1.07-19.26) 0.041
PR status
 PR+ 1 (Reference) 1 (Reference)
 PR– 4.81 (1.91-12.11) 0.001 4.92 (1.87-12.93) 0.001
ERBB2 status
ERBB2+ 1.51 (0.64-3.52) 0.344
ERBB2 1 (Reference)
Pathologic stage
 Stage I 1 (Reference) 1 (Reference)
 Stage II 1.58 (0.87-2.85) 0.132 1.86 (0.99-3.47) 0.052
 Stage III 1.63 (0.76-3.50) 0.205 1.76 (0.80-3.89) 0.159
Bilaterality
 UBC 1 (Reference)
 SBBC 1.33 (0.41-4.26) 0.633
Neoadjuvant chemotherapy
 Not done 1 (Reference)
 Done 2.32 (0.56-9.63) 0.247
Adjuvant chemotherapy
 Not done 1 (Reference)
 Done 1.34 (0.75-2.41) 0.321
Adjuvant trastuzumab treatment
 Not done 1 (Reference) 1 (Reference)
 Done 2.64 (1.13-6.18) 0.025 3.00 (1.26-7.15) 0.013
Adjuvant radiotherapy
 Not done 1 (Reference)
 Done 0.64 (0.35-1.16) 0.145
LHRH agonist treatment
 Not done 1 (Reference) 1 (Reference)
 Done 1.47 (0.78-2.79) 0.238 2.45 (1.23-4.86) 0.011
Extended hormone therapy
 Not done 1 (Reference) 1 (Reference)
 Done 0.59 (0.32-1.11) 0.101 0.45 (0.23-0.85) 0.015

CI, confidence interval; ER, estrogen receptor; HR, hazard ratio; IDC, invasive ductal carcinoma; ILC, invasive lobular carcinoma; LHRH, luteinizing hormone-releasing hormone; PR, progesterone receptor; RFSt, recurrence-free survival censored following tamoxifen discontinuation; SBBC, synchronous bilateral breast cancer; UBC, unilateral breast cancer.

  • 1. Schuurman TN, Witteveen PO, van der Wall E, Passier JL, Huitema AD, Amant F, et al. Tamoxifen and pregnancy: an absolute contraindication? Breast Cancer Res Treat. 2019;175:17–25. ArticlePubMedPDF
  • 2. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365:1687–717. ArticlePubMed
  • 3. Lash TL, Lien EA, Sorensen HT, Hamilton-Dutoit S. Genotype-guided tamoxifen therapy: time to pause for reflection? Lancet Oncol. 2009;10:825–33. ArticlePubMedPMC
  • 4. Dezentje VO, Guchelaar HJ, Nortier JW, van de Velde CJ, Gelderblom H. Clinical implications of CYP2D6 genotyping in tamoxifen treatment for breast cancer. Clin Cancer Res. 2009;15:15–21. ArticlePubMedPDF
  • 5. Goetz MP, Rae JM, Suman VJ, Safgren SL, Ames MM, Visscher DW, et al. Pharmacogenetics of tamoxifen biotransformation is associated with clinical outcomes of efficacy and hot flashes. J Clin Oncol. 2005;23:9312–8. ArticlePubMed
  • 6. Schroth W, Goetz MP, Hamann U, Fasching PA, Schmidt M, Winter S, et al. Association between CYP2D6 polymorphisms and outcomes among women with early stage breast cancer treated with tamoxifen. JAMA. 2009;302:1429–36. ArticlePubMedPMC
  • 7. Goetz MP, Schaid DJ, Wickerham DL, Safgren S, Mushiroda T, Kubo M, et al. Evaluation of CYP2D6 and efficacy of tamoxifen and raloxifene in women treated for breast cancer chemoprevention: results from the NSABP P1 and P2 clinical trials. Clin Cancer Res. 2011;17:6944–51. PubMedPMC
  • 8. Regan MM, Leyland-Jones B, Bouzyk M, Pagani O, Tang W, Kammler R, et al. CYP2D6 genotype and tamoxifen response in postmenopausal women with endocrine-responsive breast cancer: the breast international group 1-98 trial. J Natl Cancer Inst. 2012;104:441–51. PubMedPMC
  • 9. Sanchez-Spitman A, Dezentje V, Swen J, Moes D, Bohringer S, Batman E, et al. Tamoxifen pharmacogenetics and metabolism: results from the prospective CYPTAM study. J Clin Oncol. 2019;37:636–46. ArticlePubMed
  • 10. Helland T, Henne N, Bifulco E, Naume B, Borgen E, Kristensen VN, et al. Serum concentrations of active tamoxifen metabolites predict long-term survival in adjuvantly treated breast cancer patients. Breast Cancer Res. 2017;19:125.ArticlePubMedPMCPDF
  • 11. Saladores P, Murdter T, Eccles D, Chowbay B, Zgheib NK, Winter S, et al. Tamoxifen metabolism predicts drug concentrations and outcome in premenopausal patients with early breast cancer. Pharmacogenomics J. 2015;15:84–94. ArticlePubMedPMCPDF
  • 12. Almeida T, Schroth W, Nardin J, Murdter TE, Winter S, Picolotto S, et al. (Z)-Endoxifen and early recurrence of breast cancer: an explorative analysis in a prospective Brazilian study. J Pers Med. 2022;12:511.ArticlePubMedPMC
  • 13. Madlensky L, Natarajan L, Tchu S, Pu M, Mortimer J, Flatt SW, et al. Tamoxifen metabolite concentrations, CYP2D6 genotype, and breast cancer outcomes. Clin Pharmacol Ther. 2011;89:718–25. ArticlePubMedPMC
  • 14. Sanchez-Spitman AB, Moes DA, Swen JJ, Dezentje VO, Lambrechts D, Neven P, et al. Exposure-response analysis of endoxifen serum concentrations in early-breast cancer. Cancer Chemother Pharmacol. 2020;85:1141–52. ArticlePubMedPMCPDF
  • 15. Woo HI, Lee SK, Kim J, Kim SW, Yu J, Bae SY, et al. Variations in plasma concentrations of tamoxifen metabolites and the effects of genetic polymorphisms on tamoxifen metabolism in Korean patients with breast cancer. Oncotarget. 2017;8:100296–311. ArticlePubMedPMC
  • 16. Braal CL, Jager A, Hoop EO, Westenberg JD, Lommen K, de Bruijn P, et al. Therapeutic drug monitoring of endoxifen for tamoxifen precision dosing: feasible in patients with hormone-sensitive breast vancer. Clin Pharmacokinet. 2022;61:527–37. ArticlePubMedPMCPDF
  • 17. Fox P, Balleine RL, Lee C, Gao B, Balakrishnar B, Menzies AM, et al. Dose escalation of tamoxifen in patients with low endoxifen level: evidence for therapeutic drug monitoring: the TADE study. Clin Cancer Res. 2016;22:3164–71. ArticlePubMedPDF
  • 18. Love RR, Desta Z, Flockhart D, Skaar T, Ogburn ET, Ramamoorthy A, et al. CYP2D6 genotypes, endoxifen levels, and disease recurrence in 224 Filipino and Vietnamese women receiving adjuvant tamoxifen for operable breast cancer. Springerplus. 2013;2:52.ArticlePubMedPMCPDF
  • 19. Byeon JY, Kim YH, Lee CM, Kim SH, Chae WK, Jung EH, et al. CYP2D6 allele frequencies in Korean population, comparison with East Asian, Caucasian and African populations, and the comparison of metabolic activity of CYP2D6 genotypes. Arch Pharm Res. 2018;41:921–30. ArticlePubMedPDF
  • 20. Jager NG, Rosing H, Linn SC, Schellens JH, Beijnen JH. Importance of highly selective LC-MS/MS analysis for the accurate quantification of tamoxifen and its metabolites: focus on endoxifen and 4-hydroxytamoxifen. Breast Cancer Res Treat. 2012;133:793–8. ArticlePubMedPMC
  • 21. Khor CC, Winter S, Sutiman N, Murdter TE, Chen S, Lim JSL, et al. Cross-ancestry genome-wide association study defines the extended CYP2D6 locus as the principal genetic determinant of endoxifen plasma concentrations. Clin Pharmacol Ther. 2023;113:712–23. PubMed
  • 22. Mueller-Schoell A, Klopp-Schulze L, Schroth W, Murdter T, Michelet R, Brauch H, et al. Obesity alters endoxifen plasma levels in young breast cancer patients: a pharmacometric simulation approach. Clin Pharmacol Ther. 2020;108:661–70. ArticlePubMedPDF
  • 23. Puszkiel A, Arellano C, Vachoux C, Evrard A, Le Morvan V, Boyer JC, et al. Factors affecting tamoxifen metabolism in patients with breast cancer: preliminary results of the French PHACS study. Clin Pharmacol Ther. 2019;106:585–95. ArticlePubMedPDF
  • 24. Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, et al. Breast cancer, version 3.2020, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2020;18:452–78. PubMed
  • 25. Goetz MP, Sangkuhl K, Guchelaar HJ, Schwab M, Province M, Whirl-Carrillo M, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2D6 and tamoxifen therapy. Clin Pharmacol Ther. 2018;103:770–7. ArticlePubMedPMCPDF
  • 26. Puszkiel A, Arellano C, Vachoux C, Evrard A, Le Morvan V, Boyer JC, et al. Model-based quantification of impact of genetic polymorphisms and co-medications on pharmacokinetics of tamoxifen and six metabolites in breast cancer. Clin Pharmacol Ther. 2021;109:1244–55. ArticlePubMedPDF
  • 27. Martinez de Duenas E, Ochoa Aranda E, Blancas Lopez-Barajas I, Ferrer Magdalena T, Bandres Moya F, Chicharro Garcia LM, et al. Adjusting the dose of tamoxifen in patients with early breast cancer and CYP2D6 poor metabolizer phenotype. Breast. 2014;23:400–6. ArticlePubMed
  • 28. Howlader N, Cronin KA, Kurian AW, Andridge R. Differences in breast cancer survival by molecular subtypes in the United States. Cancer Epidemiol Biomarkers Prev. 2018;27:619–26. ArticlePubMedPDF
  • 29. El Saghir NS, Seoud M, Khalil MK, Charafeddine M, Salem ZK, Geara FB, et al. Effects of young age at presentation on survival in breast cancer. BMC Cancer. 2006;6:194.PubMedPMC
  • 30. Assi HA, Khoury KE, Dbouk H, Khalil LE, Mouhieddine TH, El Saghir NS. Epidemiology and prognosis of breast cancer in young women. J Thorac Dis. 2013;5 Suppl 1:S2–8. PubMed

Figure & Data

REFERENCES

    Citations

    Citations to this article as recorded by  

      • PubReader PubReader
      • ePub LinkePub Link
      • Cite
        CITE
        export Copy Download
        Close
        Download Citation
        Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

        Format:
        • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
        • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
        Include:
        • Citation for the content below
        Endoxifen Concentration Is Associated with Recurrence-Free Survival in Hormone-Sensitive Breast Cancer Patients
        Cancer Res Treat. 2025;57(1):140-149.   Published online June 18, 2024
        Close
      • XML DownloadXML Download
      Endoxifen Concentration Is Associated with Recurrence-Free Survival in Hormone-Sensitive Breast Cancer Patients
      Image Image Image
      Fig. 1. Study design.
      Fig. 2. (A) Distribution of endoxifen concentration in our study population. Kaplan-Meier curve of RFSt (B) and RFSc (C) probability of breast cancer patients according to an endoxifen cutoff of 21.00 ng/mL. RFSc, complete recurrence-free survival; RFSt, recurrence-free survival censored following tamoxifen discontinuation.
      Fig. 3. Kaplan-Meier curve of RFSt (A, C) and RFSc (B, D) probability of breast cancer patients according to an endoxifen cutoff of 5.97 ng/mL (15.98 nM) (A, B) and 10.30 ng/mL (27.58 nM) (C, D). RFSc, complete recurrence-free survival; RFSt, recurrence-free survival censored following tamoxifen discontinuation.
      Endoxifen Concentration Is Associated with Recurrence-Free Survival in Hormone-Sensitive Breast Cancer Patients
      Characteristic Total (n=478) Low endoxifen (n=218) High endoxifen (n=260) p-valuea)
      Age at diagnosis (yr) 43.8 (40.2-46.8) 43.4 (40.0-46.5) 44.0 (40.4-47.0) 0.280
      Histologic finding
       IDC 397 (83.1) 182 (83.5) 215 (82.7) 0.298
       ILC 19 (4.0) 12 (5.5) 7 (2.7)
       Mixed 28 (5.9) 10 (4.6) 18 (6.9)
       Others 34 (7.1) 14 (6.4) 20 (7.7)
      ER status
       ER+ 473 (99.0) 215 (98.6) 258 (99.2) 0.661
       ER– 5 (1.0) 3 (1.4) 2 (0.8)
      PR status
       PR+ 464 (97.1) 209 (95.9) 255 (98.1) 0.175
       PR– 14 (2.9) 9 (4.1) 5 (1.9)
      ERBB2 status
      ERBB2+ 38 (7.9) 17 (7.8) 21 (8.1) > 0.99
      ERBB2 440 (92.1) 201 (92.2) 239 (91.9)
      Pathologic stage
       Stage I 233 (48.7) 96 (44.0) 137 (52.7) 0.112
       Stage II 181 (37.9) 87 (39.9) 94 (36.2)
       Stage III 64 (13.4) 35 (16.1) 29 (11.2)
      Bilaterality
       UBC 460 (96.2) 213 (97.7) 247 (95.0) 0.153
       SBBC 18 (3.8) 5 (2.3) 13 (5.0)
      Neoadjuvant chemotherapy
       Not done 473 (99.0) 217 (99.5) 256 (98.5) 0.382
       Done 5 (1.0) 1 (0.5) 4 (1.5)
      Adjuvant chemotherapy
       Not done 187 (39.1) 80 (36.7) 107 (41.2) 0.347
       Done 291 (60.9) 138 (63.3) 153 (58.8)
      Adjuvant trastuzumab treatment
       Not done 455 (95.2) 206 (94.5) 249 (95.8) 0.531
       Done 23 (4.8) 12 (5.5) 11 (4.2)
      Adjuvant radiotherapy
       Not done 105 (22.0) 53 (24.3) 52 (20.0) 0.267
       Done 373 (78.0) 165 (75.7) 208 (80.0)
      LHRH agonist treatment
       Not done 398 (83.3) 191 (87.6) 207 (79.6) 0.029
       Done 80 (16.7) 27 (12.4) 53 (20.4)
      Extended hormone therapy
       Not done 344 (72.0) 157 (72.0) 187 (71.9) > 0.99
       Done 134 (28.0) 61 (28.0) 73 (28.1)
      Characteristic Univariable analysis
      Multivariable analysis
      HR (95% CI) p-value HR (95% CI) p-value
      Endoxifen
       Low (≤ 21.00 ng/mL) 1.78 (1.04-3.04) 0.034 1.78 (1.03-3.08) 0.038
       High (> 21.00 ng/mL) 1 (Reference) 1 (Reference)
      Age
       Per year 0.97 (0.92-1.01) 0.165
      Histologic finding
       IDC 1 (Reference)
       ILC 2.29 (0.82-6.37) 0.113
       Mixed 0.33 (0.05-2.41) 0.276
       Others 1.40 (0.55-3.53) 0.476
      ER status
       ER+ 1 (Reference) 1 (Reference)
       ER– 4.53 (1.10-18.65) 0.036 4.53 (1.07-19.26) 0.041
      PR status
       PR+ 1 (Reference) 1 (Reference)
       PR– 4.81 (1.91-12.11) 0.001 4.92 (1.87-12.93) 0.001
      ERBB2 status
      ERBB2+ 1.51 (0.64-3.52) 0.344
      ERBB2 1 (Reference)
      Pathologic stage
       Stage I 1 (Reference) 1 (Reference)
       Stage II 1.58 (0.87-2.85) 0.132 1.86 (0.99-3.47) 0.052
       Stage III 1.63 (0.76-3.50) 0.205 1.76 (0.80-3.89) 0.159
      Bilaterality
       UBC 1 (Reference)
       SBBC 1.33 (0.41-4.26) 0.633
      Neoadjuvant chemotherapy
       Not done 1 (Reference)
       Done 2.32 (0.56-9.63) 0.247
      Adjuvant chemotherapy
       Not done 1 (Reference)
       Done 1.34 (0.75-2.41) 0.321
      Adjuvant trastuzumab treatment
       Not done 1 (Reference) 1 (Reference)
       Done 2.64 (1.13-6.18) 0.025 3.00 (1.26-7.15) 0.013
      Adjuvant radiotherapy
       Not done 1 (Reference)
       Done 0.64 (0.35-1.16) 0.145
      LHRH agonist treatment
       Not done 1 (Reference) 1 (Reference)
       Done 1.47 (0.78-2.79) 0.238 2.45 (1.23-4.86) 0.011
      Extended hormone therapy
       Not done 1 (Reference) 1 (Reference)
       Done 0.59 (0.32-1.11) 0.101 0.45 (0.23-0.85) 0.015
      Table 1. Clinical characteristics of the study population

      Values are presented as median (IQR) or number (%). ER, estrogen receptor; IDC, invasive ductal carcinoma; ILC, invasive lobular carcinoma; IQR, interquartile range; LHRH, luteinizing hormone-releasing hormone; PR, progesterone receptor; SBBC, synchronous bilateral breast cancer; UBC, unilateral breast cancer.

      p-values were calculated with t tests and χ2 tests for numerical and categorical variables, respectively.

      Table 2. Univariable and multivariable Cox proportional hazards regression analyses for RFSt

      CI, confidence interval; ER, estrogen receptor; HR, hazard ratio; IDC, invasive ductal carcinoma; ILC, invasive lobular carcinoma; LHRH, luteinizing hormone-releasing hormone; PR, progesterone receptor; RFSt, recurrence-free survival censored following tamoxifen discontinuation; SBBC, synchronous bilateral breast cancer; UBC, unilateral breast cancer.


      Cancer Res Treat : Cancer Research and Treatment
      Close layer
      TOP