Purpose:

RET is an estrogen response gene with preclinical studies demonstrating cross-talk between the RET and estrogen receptor (ER) pathways. We investigate the role of lenvatinib, a multikinase inhibitor with potent activity against RET, in patients with metastatic breast cancer.

Patients and Methods:

Patients with advanced ER+/HER2 breast cancer were treated with lenvatinib plus letrozole in a phase Ib/II trial. Primary objectives included safety and recommended phase II dose (RP2D) determination in phase Ib, and objective response rates (ORR) in phase II dose expansion.

Results:

Sixteen patients were recruited in dose finding, where deescalating doses of lenvatinib from 20 to 14 mg were investigated. Lenvatinib 14 mg plus letrozole 2.5 mg daily was determined as RP2D. Thirty-one patients with 5 median lines of prior therapy in the metastatic setting (range, 0–11) were recruited in dose expansion. In this cohort, ORR was 23.3% [95% confidence interval (CI) 9.9%–42.3%], with median duration of response (DoR) of 6.9 months [interquartile range (IQR) 5.9 to 13.1]. Clinical benefit rate ≥6 months (CBR) was 50.0% (95% CI, 31.3%–68.7%). Similar efficacy was observed in the subgroup of 25 patients who progressed on prior CDK4/6 inhibitor therapy [ORR 20.0% (95% CI, 6.8%–40.7%), median DoR 6.9 months (IQR 5.9–13.1), and CBR 52.0% (95% CI, 31.3%–72.2%)]. Pharmacodynamic studies showed target modulation, with paired tumor biopsies indicating downregulation of RET/pERK and improved vascular normalization index.

Conclusions:

Lenvatinib plus letrozole had manageable toxicity, with target engagement and preliminary antitumor activity observed, supporting further assessment in randomized studies.

Translational Relevance

Forty-seven patients with advanced ER-positive HER2-negative breast cancer were treated with letrozole plus lenvatinib, a multikinase inhibitor with potent activity against RET, in a phase Ib/II study. Combination therapy at lenvatinib 14 mg with letrozole 2.5 mg daily was found to have a manageable toxicity profile, and showed promising efficacy in a heavily pretreated population of patients with 5 median lines of prior therapy (range, 0–11). The objective response rate was 23.3% (95% CI, 9.9%–42.3%), with a median duration of response of 6.9 months (interquartile range, 5.9–13.1) and clinical benefit rate ≥6 months of 50.0% (95% CI, 31.3%–68.7%). Efficacy was similar in a subgroup of patients despite having progressed on prior endocrine therapy plus CDK4/6 inhibitor therapy. Pharmacodynamic studies showed target engagement with paired tumor biopsies indicating downregulation of RET/pERK and improved vascular normalization index. These results warrant further assessment in a randomized study.

Hormone receptor–positive (HR+) breast cancer is the most common breast cancer subtype. In metastatic breast cancer (MBC), inhibitors of the CDK4/6 and PI3K/AKT/mTOR pathway (1), in combination with endocrine therapy (ET), have delayed use of chemotherapy in the absence of visceral crisis. CDK4/6 inhibitors combined with ET have emerged as standard-of-care first-line therapy, showing significant progression-free survival (PFS) and overall survival (OS) gains with consistent hazard ratios (HR) across drugs within the same class (2–6). Post CDK4/6 inhibitor exposure, few randomized studies have offered head-to-head comparison to identify the optimal second-line treatment strategy (7–10). In real-world practice, treatment choice is largely based upon physician preference and shared decision-making with the patient.

RET is an estrogen response gene with preclinical studies demonstrating cross-talk between the RET and estrogen receptor (ER) pathway (11). A recent study testing RET expression in 990 biopsy tissue from patients with primary breast cancer showed RET overexpression (defined by moderate to strong intensity staining on IHC) in 45.4% of hormone receptor–positive, HER2-negative tumors (12). Increased RET expression has been demonstrated in hormone-resistant breast cancer cell lines, with greater cell kill observed in combination of ET and RET inhibitor compared with single-agent ET (13). Lenvatinib is a multikinase inhibitor with potent activity against RET, vascular endothelial growth factor receptor (VEGFR), KIT, and fibroblastic growth factor receptor pathways, with FDA approval for use as monotherapy and combination therapy in thyroid, liver, endometrial, and renal cell carcinomas (14–18). Preclinical studies from our laboratory showed at least additive effects of lenvatinib with ET in HR+ breast cancer cell lines (manuscript in preparation). An investigator-initiated, single-center phase Ib/II study investigating safety and tolerability of the combination of lenvatinib plus letrozole, followed by dose expansion at recommended phase II dose (RP2D) was conducted. Primary objectives included safety and RP2D determination in phase Ib, and objective response rates (ORR) in phase II dose expansion.

Patient population

Patients ages 18 years or older, with histologically confirmed HR+/HER2-negative breast cancer, Eastern Cooperative Oncology Group (ECOG) performance status of 0–1, measurable disease according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 (19), and adequate organ function were eligible. No restrictions to prior line(s) of chemotherapy or ET were placed, and patients with prior exposure to letrozole were eligible if they progressed on letrozole more than 1 year after adjuvant treatment, or more than 6 months in the metastatic setting. Patients who were premenopausal were eligible if treated with medical ovarian suppression with postmenopausal levels of estradiol; patients with symptomatic brain metastases were excluded. Detailed inclusion and exclusion criteria are provided in Supplementary Data.

The study was conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization Good Clinical Practice Guideline and approved by the relevant regulatory and independent institutional review board. Written informed consent was obtained from enrolled patients.

Study design

This was an investigator-initiated phase Ib/II dose expansion, open-label study (ClinicalTrials.gov: NCT02562118) investigating the safety and tolerability of lenvatinib plus letrozole in patients with HR+/HER2 breast cancer at the National University Cancer Institute, Singapore (NCIS). The trial was initially intended to recruit early-stage breast cancer patients receiving neoadjuvant ET, but due to poor recruitment, was subsequently amended to enroll MBC patients after the first four patients were enrolled.

A 3 + 3 dose deescalation design was applied in phase Ib, with a standard dose of letrozole at 2.5 mg daily combined with varying doses of lenvatinib. Lenvatinib dosing commenced at 20 mg daily, with deescalation to lower doses if required. A 2-week safety run-in of lenvatinib monotherapy was administered prior to addition of letrozole (Fig. 1A). After RP2D was determined, dose expansion occurred to explore potential efficacy signals. Treatment was continued until disease progression, withdrawal of consent, or unacceptable toxicity.

Figure 1.

A, Treatment schema for patients on study. Patients were treated with single-agent lenvatinib for 2 weeks as run-in prior to combination treatment with letrozole until disease progression. Serial tumor biopsies were carried out at up to 4 timepoints, including baseline, 2 weeks after single-agent lenvatinib, 4 weeks after combination treatment, and at disease progression. B, Diagram of patient enrollment in both phase I dose escalation and phase II dose expansion. A total of 16 patients and 31 patients were enrolled, respectively.

Figure 1.

A, Treatment schema for patients on study. Patients were treated with single-agent lenvatinib for 2 weeks as run-in prior to combination treatment with letrozole until disease progression. Serial tumor biopsies were carried out at up to 4 timepoints, including baseline, 2 weeks after single-agent lenvatinib, 4 weeks after combination treatment, and at disease progression. B, Diagram of patient enrollment in both phase I dose escalation and phase II dose expansion. A total of 16 patients and 31 patients were enrolled, respectively.

Close modal

Serial tumor biopsies were carried out at up to 4 time points for translational studies, including baseline, 2 weeks after lenvatinib safety run-in, 4 weeks post combination therapy, and an optional biopsy at disease progression.

Study and efficacy assessments

Patients had vital sign measurements, physical examinations, ECOG performance status determination, full blood count, chemistry panel, electrocardiography, echocardiogram and computed tomography scan performed at baseline. Serial thyroid function and urine dipstick monitoring were performed for lenvatinib-specific toxicities including hypothyroidism and proteinuria. Toxicities were assessed using CTCAE version 4.0 (20). Frequency of assessments are described in Data Supplement.

Statistical methods

A 3 + 3 dose deescalation design was applied in phase I, followed by dose expansion at RP2D with target enrolment of at least 30 patients, based on Ahern's design assuming 80% power, one-sided α of 0.1, alternative hypothesis of an ORR of 50%, and null hypothesis of ORR <30%. Primary objectives of the study included the safety and determination of RP2D in phase Ib, and determination of ORR at RP2D in phase II dose expansion. All statistical analyses were generated using STATA Version 16.0 and performed on an intention-to-treat (ITT) basis, except for ORR and clinical benefit rate (CBR) where modified ITT was additionally performed to include only those who were evaluable. Details are included in Supplementary Data.

Lenvatinib pharmacokinetics, tumor pharmacodynamics, and molecular characterization

Pharmacokinetics (PK) modeling was conducted using a noncompartmental extravascular model for plasma with Phoenix WinNonLin Software version 8.0 (Centera). Pharmacodynamics (PD) biomarker analysis was carried out with IHC assessment of RET, pERK, Ki67, CD31, and SMA using formalin-fixed, paraffin-embedded tissue sections.

Hematoxylin and eosin–stained sections were studied for tumor adequacy, and biopsies with <5% tumor in them were excluded from analysis. Automated staining was done using Leica biosystems BOND-MAX system for the primary antibodies RET (ABCAM, EPR2871) and ERK1/2 MAPK 44/45 pERK (Cell Signaling; #9101), Ki67 (DAKO, #M7240), CD31 (Dako, #M0823), and SMA (Dako, #M0851). Semiquantitative scoring of marker expression was done as H-score (intensity times % of +cells) for RET and pERK, proliferation index % (Ki67), microvascular density (MVD), and vascular normalization index (CD31 and SMA).

MVD was calculated as the number of CD31+ vessels in the highest vascular hotspot microscopic field at 200× magnification. In case of >1 hotspots, MVD of the hotspots was averaged. In very tiny biopsy samples, the entire biopsy was used for MVD calculation. Vascular normalization index (VNI) was calculated as the percentage of dual CD31+ vessels coexpressing SMA on the pericyte layer.

The H-score for RET expression levels was categorized as negative/0(0–49), 1+(50–99), 2+(100–149), and 3+(150–300). H-score ≥50 (cat 1–3+) was considered RET-positive tumors, and H-score <50 was considered RET-negative tumors.

The mixed-effect model assuming a random intercept was implemented to examine the effect of response and time on the IHC assessments. This takes into account possible intrasubject correlation in repeated assessments at baseline, 2 weeks, and 4 weeks. Statistical evaluations were made assuming a two-sided test at the 5% level of significance.

Data availability

The data generated in this study are available within the article and its Supplementary Data files.

Patient characteristics

Forty-seven patients were enrolled and included in the safety analysis (16 subjects in dose escalation, 31 in dose expansion). The initial four patients enrolled received study treatment as neoadjuvant therapy for locally advanced disease, after which only patients with MBC were enrolled. All patients were evaluable for toxicity. All 31 patients in phase II were included for survival analysis, and median duration of follow-up for this cohort was 12.3 months. One patient was unevaluable for response due to death by suicide prior to first response assessment (Supplementary Fig. S1).

Median age at enrolment was 61 (range, 31–72) years, and median lines of prior therapy in the metastatic setting was 5 (range, 0–11). Among patients with MBC (n = 43), 70% had prior chemotherapy, 77% had prior ET plus CDK4/6 inhibitor therapy, and 12% had prior ET as single-agent therapy without combination with targeted therapy (e.g., CDK4/6 inhibitor or mTOR inhibitor) in the metastatic setting (Table 1).

Table 1.

Demographic and clinical characteristics of trial participants.

Dose escalationDose expansionOverall
Characteristics(n = 16)(n = 31)(n = 47)
Median age (range), year 62 (47–71) 58 (31–72) 61 (31–72) 
Histologic subtypea 
 Ductal 11 (69%) 24 (83%) 35 (78%) 
 Lobular 3 (19%) 1 (3%) 4 (9%) 
 Others 2 (12%) 4 (14%) 6 (13%) 
Histologic gradeb 
 1 2 (13%) 1 (4%) 3 (7%) 
 2 9 (60%) 10 (38) 19 (46%) 
 3 4 (27%) 15 (58) 19 (46%) 
Receptor status 
 ER-positive 16 (100%) 31 (100%) 47 (100%) 
 PR-positive 15 (94%) 26 (84%) 41 (87%) 
 HER2-positive 
Metastatic disease at point of enrolment 12 (75%) 31 (100%) 43 (91%) 
 Prior chemotherapy in neoadjuvant/adjuvant setting 1 (1%) 13 (42%) 14 (30%) 
 Median lines or previous treatment in metastatic setting (range) 3 (0–10) 5 (0–11) 4 (0–11) 
Prior therapies in metastatic setting 
 Chemotherapy 6/12 (50%) 24/31 (77%) 30/43 (70%) 
 ET + CDK 4/6 inhibitor 8/12 (67%) 25/31 (81%) 33/43 (77%) 
 ET + mTOR/PI3K inhibitor 3/12 (25%) 6/31 (19%) 9/43 (21%) 
 Single-agent ETc 2/12 (17%) 3/31 (10%) 5/43 (12%) 
Sites of metastatic disease 
 Lung 4 (33%) 22 (71%) 26 (60%) 
 Liver 6 (50%) 23 (74%) 29 (67%) 
 Bone 9 (75%) 25 (81%) 34 (79%) 
 Lymph nodes 5 (42%) 22 (71%) 27 (63%) 
 Soft tissue 1 (8%) 6 (19%) 7 (16%) 
 Others 3 (10%) 3 (7%) 
Dose escalationDose expansionOverall
Characteristics(n = 16)(n = 31)(n = 47)
Median age (range), year 62 (47–71) 58 (31–72) 61 (31–72) 
Histologic subtypea 
 Ductal 11 (69%) 24 (83%) 35 (78%) 
 Lobular 3 (19%) 1 (3%) 4 (9%) 
 Others 2 (12%) 4 (14%) 6 (13%) 
Histologic gradeb 
 1 2 (13%) 1 (4%) 3 (7%) 
 2 9 (60%) 10 (38) 19 (46%) 
 3 4 (27%) 15 (58) 19 (46%) 
Receptor status 
 ER-positive 16 (100%) 31 (100%) 47 (100%) 
 PR-positive 15 (94%) 26 (84%) 41 (87%) 
 HER2-positive 
Metastatic disease at point of enrolment 12 (75%) 31 (100%) 43 (91%) 
 Prior chemotherapy in neoadjuvant/adjuvant setting 1 (1%) 13 (42%) 14 (30%) 
 Median lines or previous treatment in metastatic setting (range) 3 (0–10) 5 (0–11) 4 (0–11) 
Prior therapies in metastatic setting 
 Chemotherapy 6/12 (50%) 24/31 (77%) 30/43 (70%) 
 ET + CDK 4/6 inhibitor 8/12 (67%) 25/31 (81%) 33/43 (77%) 
 ET + mTOR/PI3K inhibitor 3/12 (25%) 6/31 (19%) 9/43 (21%) 
 Single-agent ETc 2/12 (17%) 3/31 (10%) 5/43 (12%) 
Sites of metastatic disease 
 Lung 4 (33%) 22 (71%) 26 (60%) 
 Liver 6 (50%) 23 (74%) 29 (67%) 
 Bone 9 (75%) 25 (81%) 34 (79%) 
 Lymph nodes 5 (42%) 22 (71%) 27 (63%) 
 Soft tissue 1 (8%) 6 (19%) 7 (16%) 
 Others 3 (10%) 3 (7%) 

Abbreviations: ER, estrogen receptor; ET, endocrine therapy; PR, progesterone receptor.

aTwo patients with unknown histologic subtype in expansion phase.

bSix patients with histologic grade not available (1 dose escalation, 5 expansion phase).

cPatients who received single-agent endocrine therapy without ever combining with targeted agents such as CDK4/6 inhibitors or mTOR inhibitors.

Dose finding and expansion

Lenvatinib treatment commenced at dose level (DL) 1 of 20 mg daily (n = 4). Of 4 patients, 2 experienced dose-limiting toxicities (DLT) including grade 3 (G3) palmar–plantar erythrodysthesia (PPE; n = 1) and G3 proteinuria (n = 1), resulting in dose deescalation to DL-1 of 16 mg lenvatinib daily. At DL-1 (n = 6), no DLTs were observed, but all 6 patients required dose reductions, a majority (4/6) occurring within 6 weeks of initiation of combination therapy (G3 hypertension (3/6) and G3 wound pain (1/6)), deeming DL-1 intolerable. Six patients were enrolled at DL-2 of lenvatinib 14 mg daily and none required dose reduction, thus DL-2 was declared as RP2D. Phase II dose expansion occurred at RP2D, with 31 patients enrolled (Fig. 1B).

Safety and toxicity

Dose finding cohort

In dose-finding phase, the most common all-grade treatment-emergent adverse events (TEAE) included hypothyroidism (75%), hypertension (62.5%), proteinuria (62.5%), and PPE (62.5%; Table 2), which were deemed treatment related. The most common grade 3/4 toxicities were hypertension (43.8%), proteinuria (18.8%), and PPE (18.8%), which were managed with medication and resolved without sequelae.

Table 2.

Frequency of TEAEs.

Phase of studyDose escalationDose expansion
Dose20 mg16 mg14 mg14 mg
(n = 4)(n = 6)(n = 6)(n = 31)
Adverse eventAll gradesGrade 3/4All gradesGrade 3/4All gradesGrade 3/4All gradesGrade 3/4
Hypertensiona 20 14 
Hypothyroida 12 
Fatigue/lethargya 12 
Proteinuriaa 
Palmer–plantar erythrodysthesiaa 
Mucositisa 
Diarrhea 
Transaminitis 
Infection 
Anorexia 
Arthralgia 
Pain 
Weight loss 
Hypokalemia 
Vomiting 
Rash 
Nausea 
Hyponatremia 
Phase of studyDose escalationDose expansion
Dose20 mg16 mg14 mg14 mg
(n = 4)(n = 6)(n = 6)(n = 31)
Adverse eventAll gradesGrade 3/4All gradesGrade 3/4All gradesGrade 3/4All gradesGrade 3/4
Hypertensiona 20 14 
Hypothyroida 12 
Fatigue/lethargya 12 
Proteinuriaa 
Palmer–plantar erythrodysthesiaa 
Mucositisa 
Diarrhea 
Transaminitis 
Infection 
Anorexia 
Arthralgia 
Pain 
Weight loss 
Hypokalemia 
Vomiting 
Rash 
Nausea 
Hyponatremia 

Note: TEAEs of at least 10% all-grade frequency overall are listed.

aTEAEs deemed treatment related.

Dose expansion cohort

In dose expansion, the most common all-grade TEAEs were hypertension [64.5% (grade 3/4, 45.2%)], hypothyroidism [38.7% (grade 3/4, 0%)], and fatigue [38.7% (grade 3/4, 6.5%); Table 2]. Toxicities were deemed treatment related and consistent with lenvatinib toxicity profile, and were managed by standard guidelines. One patient experienced grade 4 hyponatremia that was deemed unrelated to study procedures, and no grade 5 TEAEs were observed. Median time for normalization of thyroid function after commencement of thyroxine for hypothyroidism was 12.6 weeks (range, 1.6–26.4 weeks). There was variable practice between investigators in the management of thyroid function post discontinuation of lenvatinib, with some continuing thyroxine replacement without attempting to taper, whereas others actively reduced the dose to facilitate weaning. At the time of data analysis, 2 patients were weaned off thyroxine at 3.6 and 32.9 weeks, respectively, after discontinuation of lenvatinib. Hypothyroidism is a known class effect of tyrosine kinase inhibitors that affect the VEGFR pathway, and studies looking at long term-effects have shown varying levels of recovery, ranging from complete resolution to permanent hypothyroidism if there is destruction of thyroid cells with thyroid gland atrophy (21, 22).

Antitumor responses and survival analyses

Antitumor activity of letrozole plus lenvatinib is detailed in Table 3 and Fig. 2. All patients (n = 16) in the dose-finding and 30 of 31 patients in the dose-expansion cohorts were evaluable for treatment response. The key primary endpoint for efficacy was measured based on ORR in the dose-expansion cohort.

Table 3.

Best tumor response by dose and phase of study.

PhaseDose escalationDose expansion, 14 mg
Dose20 mg16 mg14 mgTotalTotalPrior CDK4/6i
(n = 4)(n = 6)(n = 6)(n = 16)(n = 30a)(n = 25)
CR 0 (0%) 0 (0%) 0 (0%) 0 (0%) 0 (0%) 0 (0%) 
PR 2 (50%) 5 (83%) 2 (33%) 9 (56%) 7 (23%) 5 (20%) 
SD 2 (50%) 0 (0%) 4 (67%) 6 (38%) 12 (40%) 10 (40%) 
PD 0 (0%) 1 (17%) 0 (%) 1 (6%) 11 (37%) 10 (40%) 
ORR (95% CI) 50.0 (6.8–93.2) 83.3 (35.9–99.6) 33.3 (4.3–77.7) 56.3 (29.9–80.2) 23.3 (9.9–42.3) 20.0 (6.8–40.7) 
CBR (95% CI) 100.0 (39.8–100.0) 83.3 (35.9–99.6) 66.7 (22.3–95.7) 81.3 (54.4–96.0) 50.0 (31.3–68.7) 52.0 (31.3–72.2) 
PhaseDose escalationDose expansion, 14 mg
Dose20 mg16 mg14 mgTotalTotalPrior CDK4/6i
(n = 4)(n = 6)(n = 6)(n = 16)(n = 30a)(n = 25)
CR 0 (0%) 0 (0%) 0 (0%) 0 (0%) 0 (0%) 0 (0%) 
PR 2 (50%) 5 (83%) 2 (33%) 9 (56%) 7 (23%) 5 (20%) 
SD 2 (50%) 0 (0%) 4 (67%) 6 (38%) 12 (40%) 10 (40%) 
PD 0 (0%) 1 (17%) 0 (%) 1 (6%) 11 (37%) 10 (40%) 
ORR (95% CI) 50.0 (6.8–93.2) 83.3 (35.9–99.6) 33.3 (4.3–77.7) 56.3 (29.9–80.2) 23.3 (9.9–42.3) 20.0 (6.8–40.7) 
CBR (95% CI) 100.0 (39.8–100.0) 83.3 (35.9–99.6) 66.7 (22.3–95.7) 81.3 (54.4–96.0) 50.0 (31.3–68.7) 52.0 (31.3–72.2) 

Abbreviations: CBR, clinical benefit rate; CI, confidence interval; CR, complete response; ORR, objective response rate; PD, progressive disease; PR, partial response; SD, stable disease.

aResponse was not evaluable in one patient.

Figure 2.

Clinical outcomes of patients on study. A, Swimmer plot indicating treatment duration of patients with metastatic disease on study. B and C, Kaplan–Meier curve representing PFS (B) and OS (C) of patients treated at phase II dose expansion (blue) and subgroup of patients in dose expansion with prior CDK4/6 inhibitor therapy (red). Both show similar PFS.

Figure 2.

Clinical outcomes of patients on study. A, Swimmer plot indicating treatment duration of patients with metastatic disease on study. B and C, Kaplan–Meier curve representing PFS (B) and OS (C) of patients treated at phase II dose expansion (blue) and subgroup of patients in dose expansion with prior CDK4/6 inhibitor therapy (red). Both show similar PFS.

Close modal

Dose-finding cohort

ORR in the dose-finding cohort was 56.3% (95% CI, 29.9%–80.2%). Among patients with MBC (n = 12), 5 of 6 (83%) achieved RECIST partial response (PR) at DL-1 (16 mg lenvatinib; Table 3), despite all 6 patients requiring dose reductions of lenvatinib to 10 or 14 mg daily. Patient LL-006 is a patient with de novo MBC who commenced on study after progressing on first-line paclitaxel. She commenced on lenvatinib with letrozole as first-line ET and has ongoing treatment response of 32.7 months despite 2 dose reductions to 10 mg of lenvatinib. Another patient LL-010 with de novo MBC who progressed on 3 prior lines of ET, including CDK4/6 inhibitor, achieved ongoing PR of 25.6 months on treatment despite dose reduction to 14 mg lenvatinib since week 11 of combination therapy.

At DL-2 (14 mg lenvatinib), 2 of 6 patients achieved PR and 4 patients achieved SD as best response, with 2 patients having SD of more than 6 months (SD ≥ 6 months). A patient with 5 median prior lines of therapy, including ET, CDK4/6 inhibitors, and chemotherapy, achieved sustained SD for 15.5 months.

Dose-expansion cohort

In the dose-expansion cohort, response was observed in 7 of 30 evaluable patients, with ORR of 23.3% (95% CI, 9.9%–42.3%; Table 3). Median duration of response (DoR) was 6.9 months (IQR 5.9–13.1). Additionally, 8 patients had SD ≥ 6 months, giving a CBR of 50.0% (95% CI, 31.3%–68.7%). Twenty-two of 31 patients had progressed, with median time-to-progression of 6.2 months (IQR, 2.1–10.8; Fig. 2). At the time of data cutoff, 14 patients have passed on, and 1-year OS was 59.7% (95% CI, 38.1%–75.9%). Similar estimates were observed in the ITT population, with ORR and CBR of 22.6% (95% CI, 9.6%–41.1%) and 48.5% (95% CI, 30.2%–66.9%), respectively.

Efficacy of letrozole plus lenvatinib in CDK4/6 inhibitor–resistant MBC

In the dose-expansion cohort, 25 patients had prior ET plus CDK4/6 inhibitor treatment. Among these patients, 5 achieved PR (ORR 20.0%; 95% CI, 6.8%–40.7%), and median DoR was 6.9 months (IQR 5.9–13.1 months). Ten patients achieved SD as best response, of whom 8 achieved SD ≥ 6 months, resulting in a CBR of 52.0%. In this subgroup of 25 patients deemed to have CDK4/6 inhibitor resistance, median PFS was 6.2 months (95% CI, 2.1–10.8 months), and 1-year OS rate was 62.4% (95% CI, 37.1%–79.9%; Fig. 2). Among these 25 patients, 8 received CDK4/6 inhibitor plus ET as immediate prior therapy before trial enrolment. Interestingly, among these 8 patients, 5 demonstrated clinical benefit with letrozole plus lenvatinib [PR (n = 2), SD ≥6 months (n = 3)], yielding CBR of 62.5%. Presently, 4 patients remain on trial with continued benefit. Only 3 patients in the dose-expansion cohort received prior single-agent ET therapy without targeted therapy. These 3 patients had a median of 5 prior lines of therapy in the metastatic setting (range, 2–6), with at least 2 prior lines of ET before study enrolment. Among the three patients, no responses were observed, and median PFS was 3.91 months, with a lower 95% confidence limit of 1.61 months. However, the upper 95% confidence limited cannot be estimated.

Pharmacokinetics

Lenvatinib PK studies using blood samples collected during monotherapy run-in and combination therapy showed no significant differences in Cmax or peak-to-trough ratios, indicating that the overall PK profile of lenvatinib was similar in single-agent or combination treatment, with no significant interaction between lenvatinib and letrozole (Supplementary Fig. S2).

Tumor pharmacodynamics changes in serial tumor biopsies during lenvatinib treatment

Of the 47 patients, 38 baseline biopsies were available for IHC biomarker evaluation, of whom 30 patients had paired samples at baseline and post 2 weeks of single-agent lenvatinib (Fig. 3A). Twenty-seven patients had a tumor biopsy after 4 weeks of combination therapy. After 2 weeks of lenvatinib monotherapy, significant downregulation of RET (mean H-score change −15.6; 95% CI, −29.2 to −2.0, P = 0.026) and numerical downregulation of pERK1/2 (mean H-score change −14.6; 95% CI, −31.0 to 1.8, P = 0.080) was observed, suggesting on-target effect of the RET pathway. Surrogate markers for VEGFR pathway including MVD and VNI were studied, and we observed significant decrease in mean MVD (mean H-score change −8.4; 95% CI, −15.4 to −1.5, P = 0.019) and increase in VNI (mean H-score change 9.0; 95% CI, 3.9–14.1, P = 0.001) after 2 weeks of single-agent lenvatinib (Supplementary Table S1). Taken together, these suggest target engagement of lenvatinib and effect on downstream pathways.

Figure 3.

A, Representative picture of IHC expression staining on serial tumor biopsies. Representative photomicrograph of IHC markers showing high (top) and low (bottom) expression. Top: (a) cytoplasmic RET expression, (b) high proliferation index seen by Ki67, (c) strong cytoplasmic and membranous pERK expression, (d and e) dual IHC showing tumor microvasculature lined by CD31+ endothelial cells, for which MVD (black arrow) was used to quantify; and brown stained SMA + pericytes showing “normalized” vessels counted as VNI (green arrow). Bottom: (f–j) showing low or negative expression of markers in the same order. B, Comparison of mean scores of IHC markers between good (blue) and poor (orange) responders. Five different IHC markers were studied, including RET, pERK1/2, Ki67, microvascular density (MVD), and VNI. P values for IHC marker in each subgroup was calculated using the mixed-effect model looking at the trend of change over 3 time points.

Figure 3.

A, Representative picture of IHC expression staining on serial tumor biopsies. Representative photomicrograph of IHC markers showing high (top) and low (bottom) expression. Top: (a) cytoplasmic RET expression, (b) high proliferation index seen by Ki67, (c) strong cytoplasmic and membranous pERK expression, (d and e) dual IHC showing tumor microvasculature lined by CD31+ endothelial cells, for which MVD (black arrow) was used to quantify; and brown stained SMA + pericytes showing “normalized” vessels counted as VNI (green arrow). Bottom: (f–j) showing low or negative expression of markers in the same order. B, Comparison of mean scores of IHC markers between good (blue) and poor (orange) responders. Five different IHC markers were studied, including RET, pERK1/2, Ki67, microvascular density (MVD), and VNI. P values for IHC marker in each subgroup was calculated using the mixed-effect model looking at the trend of change over 3 time points.

Close modal

Correlative studies of IHC and treatment response

Baseline IHC studies

We first studied the association between baseline tumor RET expression and subsequent clinical benefit from letrozole plus lenvatinib. Of the 38 patients recruited in phase Ib/II with available baseline biopsies, 37 of 38 were evaluable for clinical response. Twenty-four of 37 (64.9%) and 13/37 (35.1%) patients, respectively, had RET-positive (IHC H-score categories, 1–3+) and RET-negative (IHC H-score category, 0) tumors at baseline. Twenty-two of 37 patients (59.5%) were good responders, defined as patients who achieved PR or SD ≥ 6 months. Although more patients with RET-positive tumors achieved a good response compared with those with RET-negative tumors at baseline (67% vs. 46%, P = 0.19), this difference was not statistically significant.

Serial changes

We next studied serial changes in RET pathway signaling using biopsies across 3 time points. A total of 37, 29, and 26 tumor samples were collected at baseline, 2 weeks after lenvatinib monotherapy, and 4 weeks after combination therapy, respectively, and included in this analysis (Fig. 3B).

No significant differences in baseline mean H-scores of tumor RET and pERK were observed in good compared with poor responders (RET 82.6 vs. 52.7, P = 0.129; pERK 59.3 vs. 37.9, P = 0.289). Changes in RET and pERK IHC over time with treatment were similar in both groups, likely indicating on-target effect of treatment regardless of RET or pERK expression. Good responders had decreased tumor Ki67 in response to treatment, compared with poor responders whose tumors demonstrated progressive increase in Ki67 over time. Interestingly, a significant increase in VNI was seen in good responders (P < 0.001) but not poor responders (P = 0.078). In concordance, a downward trend in MVD at the threshold of statistical significance (P = 0.082) was noted among good responders but not poor responders. Both VNI and MVD are markers of vascular normalization and taken together suggest more prominent target engagement and PD effect of lenvatinib in clinical responders than in nonresponders (Fig. 3B).

In this study, we demonstrated that the novel combination of lenvatinib and letrozole at biologically effective doses resulted in durable clinical benefit in patients with MBC with a manageable toxicity profile, including those who previously demonstrated resistance to CDK4/6 inhibitors.

Overall, treatment-related toxicities were reversible and were known toxicities of lenvatinib such as hypertension and hypothyroidism, with minimal evidence of additive toxicity when combined with letrozole. Toxicities were effectively managed with early, proactive interventions using medical therapy where clinically indicated, such as antihypertensives and thyroxine replacement. Dose finding commenced at the dose level of lenvatinib 20 mg plus letrozole 2.5 mg daily, but dose deescalation was required due to DLT of PPE and proteinuria. Although there appears to be a dose–response relationship with a higher ORR observed among patients treated with levatinib 16 mg daily compared with 14 mg daily, lenvatinib 16 mg daily was deemed intolerable due to increased toxicity requiring frequent and early dose reductions. After careful review of the risk–benefit ratio, 14 mg lenvatinib was declared as the RP2D. Lenvatinib is currently approved by the FDA at varying doses depending on tumor subtype (14–18), ranging from 12 mg daily for hepatocellular carcinoma to 24 mg daily for thyroid cancer. The declared RP2D of lenvatinib in our study is within this range of approved dosing, and no evidence of significant interaction on PK studies was observed when letrozole was coadministered with lenvatinib. This could be explained by the fact that lenvatinib and letrozole are metabolized mainly by different cytochrome enzymes, CYP3A and CYP2A, respectively (23, 24). In terms of PD effects, tumor IHC confirmed on-target effects with downregulation of RET and pERK1/2 expression as well as a decrease in MVD and increase in VNI, indicating RET and VEGFR pathway modulation, respectively.

Tyrosine kinase inhibitors with activity against RET, including cabozantinib and vandetinib, have previously been studied in patients with MBCs in phase II trials. Single-agent cabozantinib resulted in an ORR of 13.6% (95% CI, 6%–25.7%) and a 12-week disease control rate of 46.7% (95% CI, 31.7%–61.6%) in a study involving 45 patients with MBC, of whom 96% had ER-positive breast cancer with a median of 3 prior lines of chemotherapy in the metastatic setting (25), whereas another study treated 46 patients with metastatic breast cancer with prior exposure to both anthracyclines and taxane therapy with single-agent vandetinib did not observe any objective responses (26). Combination of vandetinib with fulvestrant was further studied in patients with bone-only metastatic disease who have progressed on 1 to 2 prior lines of ET, with a CBR of 41% and median PFS of 5.8 months (27). The combination of lenvatinib and letrozole has shown evidence of clinically meaningful antitumor activity in the metastatic setting. In the phase II dose-expansion cohort, despite a median of 5 prior lines of therapy, ORR was 23.3% with a CBR of 50%. Median time-to-progression was 6.2 months, with a median DoR of 6.9 months. An encouraging 1-year OS of 59.7% was observed in this heavily pretreated population.

Although there is no standard-of-care ET in the second-line-and-beyond setting, approved ET combination therapies include everolimus with exemestane (8), and alpelisib with fulvestrant (7). In the BOLERO2 and SOLAR1 studies, combination of ET with everolimus or alpelisib yielded ORR of 9.5% and 26.6%, and median PFS of 10.6 months and 11.0 months, respectively, albeit in populations that were largely CDK4/6 inhibitor naïve. Although reported outcomes of alpelisib plus fulvestrant appear to be more favorable than everolimus plus exemestane, a majority of patients enrolled in SOLAR-1 were less heavily pretreated and received fulvestrant as ET backbone, which has demonstrated superiority to aromatase inhibitors as monotherapy. The SOLAR1 trial was also benefited by biomarker selection of patients most likely to respond to alpelisib. Despite a more heavily pretreated and biomarker-unselected patient population, the combination of letrozole plus lenvatinib investigated in our study has shown promising clinical activity comparable to that reported by BOLERO-2 and SOLAR-1. Interestingly, we noticed that 23 patients (74%) treated in the dose-expansion cohort had previously progressed on letrozole monotherapy in the metastatic setting, yet 11 of 23 (47.8%) patients demonstrated clinical benefit from letrozole plus lenvatinib. Further studies are under way to understand the mechanism of action of lenvatinib in ER+ breast cancer that have progressed on ET.

We found the activity of this combination to be highly encouraging in the subgroup of patients who previously progressed on CDK4/6 inhibitors, with 20% ORR and 52% CBR at RP2D. In a subset of 8 patients in the dose expansion cohort who progressed on CDK4/6 inhibitors immediately prior to enrolment, ORR and CBR were higher at 25.0% and 62.5%, respectively, with a median PFS of 6.2 months. In a recently presented study on the use of alpelisib plus fulvestrant after progression on ET plus CDK4/6 inhibitor, ORR of 21%, CBR of 42% with median PFS of 7.3 months, and 6-month PFS of 50.4% were observed in PI3K-mutant tumors (10). This appears comparable with the observed efficacy of letrozole plus lenvatinib despite the lack of biomarker enrichment in our patient population. Overall, this supports further exploration of lenvatinib plus letrozole in CDK4/6 inhibitor–resistant MBC.

We carried out correlative tumor IHC studies to determine the effects of lenvatinib on known target pathways, including the RET, pERK, and VEGFR pathways. Inhibition of RET signaling can target downstream ERK signaling pathway (13), whereas inhibition of VEGFR can reduce neomicrovasculature and improve normalization of tumor vessels, which improves intratumoral drug delivery (28). Our study showed downregulation of RET expression and also its downstream pERK pathway, with IHC studies showing corresponding decrease in pERK IHC expression with treatment. This is similar to a pilot study (n = 10) done in patients with hormone receptor–positive, HER2-negative breast cancer treated with 10 days of vandetinib, another RET inhibitor, preoperatively, which led to significant downregulation of the pERK pathway (29). Comparison between good and poor responders to lenvatinib in our study did not reveal statistically significant differences in baseline levels or degree of change in RET expression levels on IHC, suggesting that future studies need not limit patient selection to only those with RET-positive tumors. IHC also showed evidence of inhibition of the VEGFR pathway, another known target of lenvatinib, through decreased MVD and increased VNI, indicating decreased neovascularization and normalization of vessels through recruitment of pericytes around leaky tumor vasculature. Further correlative studies may uncover potential selective biomarkers for this promising regimen.

A potential limitation of our study was that the original study design targeted to recruit patients in a neoadjuvant setting. However, due to challenges in patient recruitment, the protocol was amended to treat patients with metastatic disease instead, and thus the initial statistical assumption of 50% ORR would have been overly optimistic. We have retrospectively performed sample size estimation assuming an ORR of 10% under the null hypothesis, and an ORR of 25% under the alternative, which would have been more realistic for a population of patients with metastatic disease. In order to achieve a power of 80% with a one-sided alpha of 10%, a total of 31 patients would be required, with at least 6 responses to be considered for further investigation. In our study, we recruited 31 patients in the dose expansion cohort, of whom 30 had evaluable disease. Among these patients, 7 achieved partial response, thus meeting the minimum target for response, which is encouraging. Nonetheless, we acknowledge that this estimation was conducted post hoc and thus should be interpreted with caution.

In conclusion, lenvatinib plus letrozole achieved clinically meaningful antitumor activity that is comparable to contemporary combination endocrine therapies in the second-line-and-beyond setting with a manageable toxicity profile. Encouraging antitumor activity was observed in a subgroup of patients despite prior resistance to aromatase inhibitor monotherapy and/or prior CDK4/6 inhibitor therapy. Our results support the further development of the combination of lenvatinib and letrozole in HR+/HER2 advanced breast cancers. A randomized phase II clinical trial of lenvatinib in combination with letrozole in comparison with fulvestrant in a second-line setting is currently in development.

J.S. Lim reports personal fees from AstraZeneca, Roche, DKSH, MSD, Eisai, Novartis, and Pfizer, and grants from CTI biopharma during the conduct of the study. A.L. Wong reports other support from Otsuka Pharmaceuticals, Pfizer, Novartis, and Eisai outside the submitted work. S.G. Ow reports personal fees from Pfizer, AstraZeneca, Roche, Novartis, and Lilly during the conduct of the study. D.S. Tan reports grants and nonfinancial support from Eisai during the conduct of the study; grants, personal fees, and nonfinancial support from AstraZeneca, Eisai, and MSD, grants from National Medical Research Council Clinician Scientist Award, personal fees from GSK, grants and nonfinancial support from Roche, and grants from Pangestu Family Foundation Gynaecological Cancer Research Fund outside the submitted work. R. Sundar reports other support from Bristol-Myers Squibb, Merck, Eisai, Bayer, Taiho, Novartis, MSD, Eli Lilly, BMS, Roche, Taiho, AstraZeneca, DKSH, Roche, AstraZeneca, Taiho, and Eisai and grants from Paxman Coolers outside the submitted work. B. Tai reports other support from Boehringer Ingelheim outside the submitted work, and received royalty from Wiley-Blackwell for the publication of books. B. Goh reports grants and nonfinancial support from MSD and Adagene, and nonfinancial support from Taiho pharmaceuticals and BMS outside the submitted work. S. Lee reports grants, personal fees, and other support from Eisai during the conduct of the study; grants, personal fees, and other support from Pfizer, personal fees from Novartis, AstraZeneca, Eli Lilly, Daiichi Sankyo, and Roche, grants and personal fees from MSD, grants from Karyopharm, and other support from Taiho and ASLAN Pharmaceuticals outside the submitted work. No disclosures were reported by the other authors.

J.S.J. Lim: Data curation, formal analysis, validation, investigation, writing–original draft, writing–review and editing. A.L.A. Wong: Investigation, writing–review and editing. S.G.W. Ow: Investigation, writing–review and editing. N.Y.L. Ngoi: Investigation, writing–review and editing. G.H.J. Chan: Investigation, writing–review and editing. Y.L.E. Ang: Investigation, writing–review and editing. W.Q. Chong: Investigation, writing–review and editing. S.E. Lim: Investigation, writing–review and editing. Y.W. Lim: Investigation, writing–review and editing. M. Lee: Investigation, writing–review and editing. J.R.E. Choo: Investigation, writing–review and editing. H.L. Tan: Investigation, writing–review and editing. W.P. Yong: Investigation, writing–review and editing. R.A. Soo: Investigation, writing–review and editing. D.S.P. Tan: Investigation, writing–review and editing. C.E. Chee: Investigation, writing–review and editing. R. Sundar: Investigation, writing–review and editing. K. Yadav: Investigation, writing–review and editing. S. Jain: Data curation, formal analysis, validation, investigation, writing–original draft, writing–review and editing. L. Wang: Data curation, formal analysis, writing–review and editing. B. Tai: Data curation, software, formal analysis, validation, investigation, writing–original draft, writing–review and editing. B.C. Goh: Investigation, writing–review and editing. S.-C. Lee: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, writing–original draft, writing–review and editing.

This study was supported by the National Medical Research Council, Singapore (NMRC/CSA/015/2009, NMRC/CSA-SI/0004/2015, NMRC/CG/012/2013, NMRC/CG/M005/2017_NCIS, MOH-000414) and Eisai Pharmaceuticals. We are grateful for all patients who participated, without whom the study would not have been possible. We would also like to thank Eisai for provision of lenvatinib, and the National Medical Research Council for grant provision for manpower and study support (NMRC/CSA/015/2009, NMRC/CSA-SI/0004/2015, MOH-000414, NMRC/CG/M005/2017_NCIS, and NMRC/CG/012/2013).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Arnedos
M
,
Vicier
C
,
Loi
S
,
Lefebvre
C
,
Michiels
S
,
Bonnefoi
H
, et al
.
Precision medicine for metastatic breast cancer—limitations and solutions
.
Nat Rev Clin Oncol
2015
;
12
:
693
704
.
2.
Li
J
,
Huo
X
,
Zhao
F
,
Ren
D
,
Ahmad
R
,
Yuan
X
, et al
.
Association of cyclin-dependent kinases 4 and 6 inhibitors with survival in patients with hormone receptor-positive metastatic breast cancer: a systematic review and meta-analysis
.
JAMA Netw Open
2020
;
3
:
e2020312
.
3.
Finn
RS
,
Martin
M
,
Rugo
HS
,
Jones
S
,
Im
S-A
,
Gelmon
K
, et al
.
Palbociclib and letrozole in advanced breast cancer
.
N Engl J Med
2016
;
375
:
1925
36
.
4.
Johnston
S
,
Martin
M
,
Di Leo
A
,
Im
S-A
,
Awada
A
,
Forrester
T
, et al
.
MONARCH 3 final PFS: a randomized study of abemaciclib as initial therapy for advanced breast cancer
.
NPJ Breast Cancer
2019
;
5
:
5
.
5.
Tripathy
D
,
Im
S-A
,
Colleoni
M
,
Franke
F
,
Bardia
A
,
Harbeck
N
, et al
.
Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): a randomised phase 3 trial
.
Lancet Oncol
2018
;
19
:
904
15
.
6.
Slamon
DJ
,
Neven
P
,
Chia
S
,
Fasching
PA
,
De Laurentiis
M
,
Im
S-A
, et al
.
Phase III randomized study of ribociclib and fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: MONALEESA-3
.
J Clin Oncol
2018
;
36
:
2465
72
.
7.
André
F
,
Ciruelos
EM
,
Juric
D
,
Loibl
S
,
Campone
M
,
Mayer
IA
, et al
.
Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1
.
Ann Oncol
2021
;
32
:
208
17
.
8.
Baselga
J
,
Campone
M
,
Piccart
M
,
Burris
HA
,
Rugo
HS
,
Sahmoud
T
, et al
.
Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer
.
N Engl J Med
2012
;
366
:
520
9
.
9.
Chia
S
,
Gradishar
W
,
Mauriac
L
,
Bines
J
,
Amant
F
,
Federico
M
, et al
.
Double-blind, randomized placebo controlled trial of fulvestrant compared with exemestane after prior nonsteroidal aromatase inhibitor therapy in postmenopausal women with hormone receptor-positive, advanced breast cancer: results from EFECT
.
J Clin Oncol
2008
;
26
:
1664
70
.
10.
Rugo
HS
,
Lerebours
F
,
Ciruelos
E
,
Drullinsky
P
,
Ruiz-Borrego
M
,
Neven
P
, et al
.
Alpelisib plus fulvestrant in PIK3CA-mutated, hormone receptor-positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): one cohort of a phase 2, multicentre, open-label, non-comparative study
.
Lancet Oncol
2021
;
22
:
489
98
.
11.
Boulay
A
,
Breuleux
M
,
Stephan
C
,
Fux
C
,
Brisken
C
,
Fiche
M
, et al
.
The Ret receptor tyrosine kinase pathway functionally interacts with the ERalpha pathway in breast cancer
.
Cancer Res
2008
;
68
:
3743
51
.
12.
Mechera
R
,
Soysal
SD
,
Piscuoglio
S
,
Ng
CKY
,
Zeindler
J
,
Mujagic
E
, et al
.
Expression of RET is associated with oestrogen receptor expression but lacks prognostic significance in breast cancer
.
BMC Cancer
2019
;
19
:
41
.
13.
Spanheimer
PM
,
Park
J-M
,
Askeland
RW
,
Kulak
MV
,
Woodfield
GW
,
De Andrade
JP
, et al
.
Inhibition of RET increases the efficacy of antiestrogen and is a novel treatment strategy for luminal breast cancer
.
Clin Cancer Res
2014
;
20
:
2115
.
14.
Kudo
M
,
Finn
RS
,
Qin
S
,
Han
K-H
,
Ikeda
K
,
Piscaglia
F
, et al
.
Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial
.
Lancet
2018
;
391
:
1163
73
.
15.
Schlumberger
M
,
Tahara
M
,
Wirth
LJ
,
Robinson
B
,
Brose
MS
,
Elisei
R
, et al
.
Lenvatinib versus placebo in radioiodine-refractory thyroid cancer
.
N Engl J Med
2015
;
372
:
621
30
.
16.
Motzer
R
,
Alekseev
B
,
Rha
S-Y
,
Porta
C
,
Eto
M
,
Powles
T
, et al
.
Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma
.
N Engl J Med
2021
;
384
:
1289
300
.
17.
Makker
V
,
Taylor
MH
,
Aghajanian
C
,
Oaknin
A
,
Mier
J
,
Cohn
AL
, et al
.
Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer
.
J Clin Oncol
2020
;
38
:
2981
92
.
18.
Motzer
RJ
,
Hutson
TE
,
Glen
H
,
Michaelson
MD
,
Molina
A
,
Eisen
T
, et al
.
Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial
.
Lancet Oncol
2015
;
16
:
1473
82
.
19.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
.
New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
20.
Trotti
A
,
Colevas
A
,
Setser
A
,
Rusch
V
,
Jaques
D
,
Budach
V
, et al
.
CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment
.
Semin Radiat Oncol
2003
;
13
:
176
81
.
21.
Mannavola
D
,
Coco
P
,
Vannucchi
G
,
Bertuelli
R
,
Carletto
M
,
Casali
PG
, et al
.
A novel tyrosine-kinase selective inhibitor, sunitinib, induces transient hypothyroidism by blocking iodine uptake
.
J Clin Endocrinol Metab
2007
;
92
:
3531
4
.
22.
Sato
S
,
Muraishi
K
,
Tani
J
,
Sasaki
Y
,
Tokubuchi
I
,
Tajiri
Y
, et al
.
Clinical characteristics of thyroid abnormalities induced by sunitinib treatment in Japanese patients with renal cell carcinoma
.
Endocr J
2010
;
57
:
873
80
.
23.
Hussein
Z
,
Mizuo
H
,
Hayato
S
,
Namiki
M
,
Shumaker
R
.
Clinical pharmacokinetic and pharmacodynamic profile of lenvatinib, an orally active, small-molecule, multitargeted tyrosine kinase inhibitor
.
Eur J Drug Metab Pharmacokinet
2017
;
42
:
903
14
.
24.
Desta
Z
,
Kreutz
Y
,
Nguyen
AT
,
Li
L
,
Skaar
T
,
Kamdem
LK
, et al
.
Plasma letrozole concentrations in postmenopausal women with breast cancer are associated with CYP2A6 genetic variants, body mass index, and age
.
Clin Pharmacol Ther
2011
;
90
:
693
700
.
25.
Tolaney
SM
,
Nechushtan
H
,
Ron
I-G
,
Schöffski
P
,
Awada
A
,
Yasenchak
CA
, et al
.
Cabozantinib for metastatic breast carcinoma: results of a phase II placebo-controlled randomized discontinuation study
.
Breast Cancer Res Treat
2016
;
160
:
305
12
.
26.
Miller
KD
,
Trigo
JM
,
Wheeler
C
,
Barge
A
,
Rowbottom
J
,
Sledge
G
, et al
.
A multicenter phase II trial of ZD6474, a vascular endothelial growth factor receptor-2 and epidermal growth factor receptor tyrosine kinase inhibitor, in patients with previously treated metastatic breast cancer
.
Clin Cancer Res
2005
;
11
:
3369
76
.
27.
Clemons
MJ
,
Cochrane
B
,
Pond
GR
,
Califaretti
N
,
Chia
SKL
,
Dent
RA
, et al
.
Randomised, phase II, placebo-controlled, trial of fulvestrant plus vandetanib in postmenopausal women with bone only or bone predominant, hormone-receptor-positive metastatic breast cancer (MBC): the OCOG ZAMBONEY study
.
Breast Cancer Res Treat
2014
;
146
:
153
62
.
28.
Martin
JD
,
Seano
G
,
Jain
RK
.
Normalizing function of tumor vessels: progress, opportunities, and challenges
.
Annu Rev Physiol
2019
;
81
:
505
34
.
29.
Spanheimer
PM
,
Bashir
A
,
Lorenzen
AW
,
Beck
AC
,
Liao
J
,
Lizarraga
IM
, et al
.
A pilot study of preoperative vandetanib on markers of proliferation and apoptosis in breast cancer
.
Am J Clin Oncol
2021
;
44
:
456
62
.

Supplementary data