Assessing the effectiveness and safety of surufatinib versus everolimus or sunitinib in advanced neuroendocrine neoplasms: insights from a real-world, retrospective cohort study using propensity score and inverse probability treatment weighting analysis
Original Article

Assessing the effectiveness and safety of surufatinib versus everolimus or sunitinib in advanced neuroendocrine neoplasms: insights from a real-world, retrospective cohort study using propensity score and inverse probability treatment weighting analysis

Linhui Zhu1,2, Xuan Ye1,2, Youjun She1,2, Wensheng Liu1,2, Kiyoshi Hasegawa3, Roberta Elisa Rossi4, Qiong Du1,2, Qing Zhai1,2

1Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China; 2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; 3Hepato-Biliary and Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan; 4Gastroenterology and Endoscopy Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy

Contributions: (I) Conception and design: L Zhu, Q Zhai; (II) Administrative support: L Zhu, X Ye, Q Du; (III) Provision of study materials or patients: L Zhu, Y She; (IV) Collection and assembly of data: X Ye, W Liu; (V) Data analysis and interpretation: L Zhu, Q Du, Q Zhai; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Qiong Du, MSc; Qing Zhai, PhD. Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dong’an Road, Xuhui District, Shanghai 200032, China. Email: dujoan-88@163.com; zhaiqing63@126.com.

Background: While surufatinib, sunitinib, and everolimus have shown efficacy for advanced neuroendocrine neoplasms (NENs) in randomized controlled trials (RCTs), direct comparisons in a real-world setting remain unexplored. This gap highlights the clinical need to understand their comparative effectiveness and safety within the diverse Chinese population. Addressing this, our study provides insights into the real-world performance of these therapies, aiming to inform treatment selection and improve patient outcomes.

Methods: A retrospective, observational study was conducted at Fudan University Shanghai Cancer Center, including patients with advanced NENs treated with surufatinib, sunitinib, or everolimus between July 2020 and April 2023. Eligibility criteria focused on histologically confirmed, locally advanced, unresectable, or metastatic NENs, with patients having received at least one month of targeted therapy. We employed inverse probability weighting (IPW) with the propensity score (PS) matching to adjust for the bias of baseline characteristics. The assessment of covariates included age, sex, performance status, primary tumor site, functional status, genetic mutations, tumor differentiation, Ki67 index, tumor grade, metastasis site, and previous therapies. The primary outcome was progression-free survival (PFS), and secondary outcomes included objective response rate (ORR), disease control rate (DCR), and adverse events (AEs).

Results: The study enrolled 123, 56, and 68 locally advanced or metastatic NEN patients treated with surufatinib, sunitinib, and everolimus, respectively. Before adjusting for confounding factors, surufatinib was used less frequently as a first-line treatment compared to sunitinib and everolimus in pancreatic NENs (pNENs) (11.1% vs. 22.1%, P=0.057). Significant differences were noted in prior treatments and tumor characteristics between surufatinib and everolimus groups in extrapancreatic NENs (epNENs) (P<0.05). Post-IPW, these disparities were resolved (P>0.05). Surufatinib demonstrated superior median PFS (mPFS) in both pancreatic [8.30 vs. 6.33 months, hazard ratio (HR) 0.592, P<0.001] and epNENs (8.73 vs. 3.70 months, HR 0.608, P<0.001) compared to everolimus or sunitinib. Notably, male gender (HR 1.75, P=0.001), functional status (HR 2.09, P=0.01), Ki67 index >20% (HR 12.7, P=0.004), previous somatostatin analogue (SSA) treatment (HR 1.73, P=0.001), germline mutation (HR 5.62, P<0.001), poor differentiation (HR 7.45, P<0.001), liver metastasis (HR 1.72, P=0.001) and multiple treatment lines (HR 1.62 for 2nd line, P=0.04; HR 1.88 for ≥3rd line, P=0.01) were identified as negative prognostic factors for PFS. Conversely, dose adjustment (HR 0.63, P=0.009) and treatment with surufatinib (HR 0.58 for pNEN, P<0.001; HR 0.62 for epNEN, P=0.002) were correlated with longer PFS.

Conclusions: In a real-world Chinese cohort, surufatinib significantly outperformed sunitinib and everolimus in prolonging PFS among advanced NEN patients, with identifiable clinical features impacting survival, and conclusions regarding superiority should be interpreted with caution due to the retrospective design. Our findings underscore the need for prospective studies to further validate these results and explore additional predictive biomarkers for personalized treatment strategies.

Keywords: Neuroendocrine neoplasms (NENs); real-world study; targeted therapy; inverse probability treatment weighting; surufatinib


Submitted Mar 25, 2024. Accepted for publication Apr 17, 2024. Published online Apr 29, 2024.

doi: 10.21037/jgo-24-218


Highlight box

Key findings

• In a real-world setting in China, surufatinib significantly prolongs progression free survival for locally advanced or metastatic pancreatic neuroendocrine neoplasms (NENs) and extrapancreatic NENs patients compared with other targeted therapies.

What is known and what is new?

• Surufatinib, sunitinib and everolimus, all of which have been approved as standard treatment for advanced NENs patients in China, showed outstanding efficacy and safety compared with placebo in randomized controlled trials, while a head-to-head comparison and real-world studies are still lacking.

• This study demonstrates that the effectiveness and safety of surufatinib appears to be better than sunitinib and everolimus in Chinese real-life clinical practice. Potential predictive factors (e.g., tumor differentiation, Ki67 index, liver metastases) and clinical treatment pattern on targeted therapies have been also investigated.

What is the implication, and what should change now?

• Effectiveness data and safety profile, together with predictive clinical features, may lead to implications for selection of targeted agents in Chinese advanced NEN patients. Furthermore, these results should be investigated and validated in well-designed prospective studies, in which more laboratory and molecular biomarkers should be explored to facilitate personalized treatments for advanced NEN.


Introduction

Neuroendocrine neoplasms (NENs) represent a rare entity originating from neuroendocrine cells, accounting for about 0.5% of all newly diagnosed malignancies (1,2). In the last decades, the incidence of NENs has shown a significant upward trend worldwide (3,4), with a large-sample study (64,791 NEN cases) based on the Surveillance, Epidemiology, and End Results (SEER) database finding that the age-adjusted incidence in 2012 (6.98/100,000 people) increased 6.4 times from that in 1973 (1.09/100,000 people) (5). NENs are highly heterogeneous tumors which can be divided into well differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs) according to tumor differentiation, with significant differences in their natural course and prognosis (6).

In view of the high complexity, rapidly growing incidence, and relatively longer survival, extensive treatment demands occur in NEN patients, and long-term and personalized health management play a crucial role. Surgery is the cornerstone of NEN therapeutic strategies, but 13–44% of NEN patients are detected with metastatic diseases at the diagnosis (7,8). Systemic therapy remains vital in advanced/metastatic NEN patients to control tumor growth and alleviate symptoms, which have been continuously developed in the past few decades, including biotherapy, chemotherapy, targeted therapy, and radiopharmaceuticals (6,9). With the gradual exploration of molecular pathways in NEN, relatively less toxic and efficient targeted therapies have received promising attention. In this context, the National Comprehensive Cancer Network (NCCN), European Neuroendocrine Tumor Society (ENETS), and Chinese Society of Clinical Oncology (CSCO) guidelines recommended targeted drugs as suitable options for patients with advanced NENs (10-12). Currently, three targeted drugs have been approved in Chinese clinical practice including surufatinib, sunitinib, and everolimus.

NENs are characterized by a highly vascularized microenvironment, in which multiple angiogenic factors drive tumorigenesis and progression (13-15). Surufatinib, developed independently in China, is a novel tyrosine kinase inhibitor (TKI) which simultaneously targets vascular endothelial growth factor receptor (VEGFR), fibroblast growth factor receptor (FGFR), and colony stimulating factor-1 receptor (CSF1 receptor), and has been shown to prolong progression-free survival (PFS) in advanced pancreatic (pNEN) and extrapancreatic NEN (epNEN) through the SANET-p and SANET-ep phase III trials (16,17). At present, surufatinib covers nearly all site-originated NEN, whereas sunitinib has shown survival benefits for advanced pNEN (18) and everolimus, an oral mTOR inhibitor (mTORi), was reported to achieve a significantly longer PFS in advanced gastroenteropancreatic (GEP) or lung NENs through the RADIANT-3 and RADIANT-4 trials (19,20).

While surufatinib, sunitinib and everolimus have shown efficacy for advanced NENs in randomized controlled trials (RCTs), direct comparisons in a real-world setting remain unexplored. Due to the heterogeneity of NEN, the difficulty of diagnosis and treatment in real-world clinical practice appears significantly higher than in strictly-controlled clinical trial environments. The actual benefits of targeted drugs remain unclear, so this gap highlights the clinical need to understand their comparative effectiveness and safety within the diverse Chinese population. Therefore, we conducted an observational study and analyzed real-world data to evaluate the efficacy and safety of surufatinib compared with sunitinib and everolimus for advanced NEN patients in China, in order to provide insights, inform treatment selection and improve patient outcomes into the real-world performance. We present this article in accordance with the STROBE reporting checklist (available at https://jgo.amegroups.com/article/view/10.21037/jgo-24-218/rc).


Methods

Study design and patients

This was a retrospective cohort study conducted at Fudan University Shanghai Cancer Center. Patients treated with surufatinib, sunitinib, or everolimus between July 2020 and April 2023 were identified, with the observation period lasting from first administration of targeted agents until disease progression/death, loss to follow-up, or the end of the study. The follow-up was until November 2023. Genomic profiles for pNEN were clearer than epNEN, in which more abundant evidences demonstrated that molecular circuits regulating angiogenesis exerts a critical role throughout the development of pNEN (14,21). Furthermore, considering the differences in disease characteristics and indications, the included patients were divided into pNEN and epNEN patients, among whom the study analysis were made on surufatinib treatment cohort vs. sunitinib or everolimus (sunitinib + everolimus) treatment cohort for pNEN patients and surufatinib treatment cohort vs. everolimus treatment cohort for epNEN patients.

The eligibility criteria were as follows: histological diagnosis of locally advanced unresectable or metastatic NEN (including pheochromocytoma/paraganglioma), having already received at least 1 month of targeted therapy. Patients with incomplete medical records were excluded. During baseline and follow-up period, clinical characteristics, treatment, efficacy and safety pertaining to included patients were collected through the electronic hospital information system (HIS) and telephone, based on International Agency for Research on Cancer (IARC)/World Health Organization (WHO) classification framework and consensus (22). The sample size of the study was determined by the number of patients that could be retrospectively collected from HIS of Fudan University Shanghai Cancer Center. Clinico-pathologic variables were collected and assessed for the propensity score (PS) calculation, including age, sex, Eastern Cooperative Oncology Group performance status (ECOG-PS), primary tumor site, functional status, genetic mutations, tumor differentiation, Ki67 index, tumor grade, metastasis site, and previous therapies. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). This study was approved by institutional ethics committee of Fudan University Shanghai Cancer Center (No. 2205254-19) and individual consent for this retrospective analysis was waived.

Treatment

The standard doses were set at 300 mg for surufatinib, 37.5 mg for sunitinib, and 10 mg for everolimus, once daily continuously. Initial dose, dose modification, and dose interruption were made solely at the physician’s discretion, according to disease severity and patient tolerance. Moreover, we imposed no restrictions on treatment lines or combination therapies, leaving this to the judgement of the medical staff.

Evaluation and endpoints

The primary outcome was PFS, defined as the time from first targeted agent administration to disease progression or death. Tumor responses were evaluated routinely every 2–3 months as complete response (CR)/partial response (PR)/stable disease (SD)/progressive disease (PD) according to the Response Evaluation Criteria in Solid Tumors (RECIST, version 1.1) (23). If disease progression or death had not occurred in a certain patient, the last follow-up time was censored. Secondary outcomes included objective response rate (ORR), defined as the proportion of patients achieving CR and PR; disease control rate (DCR), defined as the proportion of patients with CR, PR, and SD; adverse events (AEs), identified and graded based on the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE, version 5.0) (24).

Statistics

Descriptive statistical methods were utilized to summarize data. The Pearson’s chi-square test/Fisher’s exact test and the Wilcoxon test/t-test were used to assess differences of categorical variables and continuous variables among groups.

Inverse probability weighting (IPW) with the PS was applied to account for the bias of baseline characteristics, which created a pseudo-population for the following analysis between targeted agent groups. IPW adjusts weights based on PSs to address confounding factors. Compared to PS matching, IPW method does not result in sample size loss and can fully utilize all case data to avoid sample selection bias. Unadjusted and IPW-adjusted Kaplan-Meier curves were drawn, and PFS was compared using Cox proportional hazards model with hazard ratio (HR) and 95% confidence interval (CI). ORR and DCR were compared with the Pearson’s chi-square test method. Univariate and multivariate analyses were performed using Cox proportional hazards model to determine meaningful independent indicators on efficacy outcomes with HR and 95% CI estimation. In addition, exploratory subgroup analyses were performed for PFS between treatment groups.

Statistical analyses were performed with SAS software, version 9.4 (SAS Institute, Cary, NC, USA). A two-sided significance level of P<0.05 on all statistic tests was defined.


Results

Patient characteristics

Out of 267 assessed patients, those eligible were enrolled as follows: 123 in the surufatinib group, 56 in the sunitinib group, and 68 in the everolimus group (Figure 1). Table 1 summarizes the patients’ baseline characteristics. The inferior baseline profile of the surufatinib group, compared to others, created an imbalance between groups, hence all were incorporated into the PS calculation for IPW. Before IPW, for pNEN, the number of patients receiving surufatinib as first-line treatment was slightly less than that in the sunitinib + everolimus group (11.1% vs. 22.1%, P=0.057); for epNEN, there were significant differences in previous somatostatin analogues (SSA), primary tumor site, liver metastasis, treatment line, and treatment regimen between surufatinib and everolimus (P<0.05). After IPW adjustment, all baseline data of the two groups were balanced without significant differences (P>0.05).

Figure 1 Flow chart of patient enrollment. pNEN, pancreatic neuroendocrine neoplasm; epNEN, extrapancreatic neuroendocrine neoplasm; IPW, inverse probability weighting.

Table 1

Patient baseline characteristics

Characteristics Unadjusted cohort IPW-adjusted cohort
pNEN epNEN pNEN epNEN
Suru, N=36 Suni + Eve, N=77 P value Suru, N=87 Eve, N=41 P value Suru Suni + Eve P value Suru Eve P value
Sex 0.72 0.43 0.72 0.91
   Male 16 (44.4) 37 (48.1) 51 (58.6) 21 (51.2) 50.2% 52.6% 50.8% 51.5%
   Female 20 (55.6) 40 (51.9) 36 (41.4) 20 (48.8) 49.8% 47.4% 49.2% 48.5%
Age, years 50.6±14.7 50.2±13.0 0.76 54.3±11.8 52.9±10.3 0.51 48.2±26.2 49.7±15.3 0.57 54.4±14.6 54.6±18.0 0.92
ECOG-PS >0.99 0.59 0.34 0.47
   0−1 36 (100.0) 76 (98.7) 85 (97.7) 39 (95.1) 100% 99.1% 96.7% 98.1%
   ≥2 0 1 (1.3) 2 (2.3) 2 (4.9) 0% 0.9% 3.3% 1.9%
Primary tumor site 0.005 0.32
   Thymus/mediastinum 28 (32.2) 6 (14.6) 25.2% 20.3%
   Lung 16 (18.4) 8 (19.5) 24.1% 34.7%
   Colorectum 13 (14.9) 16 (39.0) 22.5% 22.2%
   Stomach 4 (4.6) 3 (7.3) 5% 4.2%
   Small intestine 0 3 (7.3) 0% 2.4%
   Other 13 (14.9) 3 (7.3) 11.8% 7.6%
   Multiple primary 7 (8.0) 1 (2.4) 6.1% 4.9%
   Unknown primary 6 (6.9) 1 (2.4) 5.3% 3.7%
Functional status 0.38 >0.99 0.45 0.38
   Functional 3 (8.3) 3 (3.9) 4 (4.6) 2 (4.9) 8.1% 5.5% 5% 2.9%
   Non-functional 33 (91.7) 74 (96.1) 83 (95.4) 39 (95.1) 91.9% 94.5% 95% 97.1%
Genetic syndrome 0.55 0.14 0.10 0.68
   Sporadic 36 (100.0) 74 (96.1) 74 (85.1) 39 (95.1) 100% 97.4% 88.4% 86.7%
   Germline mutated 0 3 (3.9) 13 (14.9) 2 (4.9) 0% 2.6% 11.6% 13.3%
Differentiation 0.71 0.16
   Well-differentiated 36 (100.0) 77 (100.0) 80 (92.0) 39 (95.1) 100% 100% 92.5% 87.2%
   Poorly-differentiated 0 0 7 (8.0) 2 (4.9) 0% 0% 7.5% 12.8%
Ki67 index 0.44 0.17 0.88 0.93
   <3% 1 (2.8) 1 (1.3) 7 (8.0) 5 (12.2) 1.4% 1.3% 7.1% 6.1%
   3−20% 32 (88.9) 64 (83.1) 57 (65.5) 31 (75.6) 82.5% 85% 71.3% 71.4%
   >20% 3 (8.3) 12 (15.6) 23 (26.4) 5 (12.2) 16% 13.7% 21.6% 22.5%
Distant metastasis >0.99 0.16 0.80 0.32
   Locally advanced 1 (2.8) 2 (2.6) 9 (10.3) 1 (2.4) 3.6% 3% 7.8% 11.4%
   Distant metastatic 35 (97.2) 75 (97.4) 78 (89.7) 40 (97.6) 96.4% 97% 92.2% 88.6%
Metastatic site number 0.54 0.80 0.44 0.82
   0 33 (91.7) 66 (85.7) 64 (73.6) 31 (75.6) 90.3% 87% 75.6% 76.7%
   ≥3 3 (8.3) 11 (14.3) 23 (26.4) 10 (24.4) 9.7% 13% 24.4% 23.3%
Liver metastasis 0.67 0.02 0.80 0.48
   Yes 33 (91.7) 73 (94.8) 39 (44.8) 27 (65.9) 93.9% 94.6% 50.1% 54.6%
   No 3 (8.3) 4 (5.2) 48 (55.2) 14 (34.1) 6.1% 5.4% 49.9% 45.4%
Bone metastasis 0.88 0.54 0.19 0.41
   Yes 6 (16.7) 12 (15.6) 46 (52.9) 24 (58.5) 19.6% 13.1% 48.4% 43.2%
   No 30 (83.3) 65 (84.4) 41 (47.1) 17 (41.5) 80.4% 86.9% 51.6% 56.8%
Prior surgery 0.20 0.45 0.46 0.23
   Yes 15 (41.7) 42 (54.5) 47 (54.0) 25 (61.0) 42.7% 47.7% 53.1% 60.5%
   No 21 (58.3) 35 (45.5) 40 (46.0) 16 (39.0) 57.3% 52.3% 46.9% 39.5%
Prior radiotherapy >0.99 0.51 0.30 0.28
   Yes 0 1 (1.3) 26 (29.9) 10 (24.4) 0% 1% 32% 38.5%
   No 36 (100.0) 76 (98.7) 61 (70.1) 31 (75.6) 100% 99% 68% 61.5%
Previous SSA 0.40 <0.001 0.42 0.59
   Yes 24 (66.7) 45 (58.4) 26 (29.9) 26 (63.4) 65.7% 60.4% 41.9% 38.6%
   No 12 (33.3) 32 (41.6) 61 (70.1) 15 (36.6) 34.3% 39.6% 58.1% 61.4%
Previous anti-ang agents 0.13 0.07 0.56 0.83
   Yes 6 (16.7) 23 (29.9) 12 (13.8) 11 (26.8) 21.7% 25.1% 20.6% 21.7%
   No 30 (83.3) 54 (70.1) 75 (86.2) 30 (73.2) 78.3% 74.9% 79.4% 78.3%
Previous mTORi 0.16 0.34 0.28 0.93
   Yes 1 (2.8) 9 (11.7) 7 (8.0) 6 (14.6) 5% 8.8% 8.3% 8.6%
   No 35 (97.2) 68 (88.3) 80 (92.0) 35 (85.4) 95% 91.2% 91.7% 91.4%
Previous chemotherapy 0.12 0.3 0.93 0.27
   Yes 16 (44.4) 46 (59.7) 53 (60.9) 21 (51.2) 55.3% 55.8% 58.5% 65.2%
   No 20 (55.6) 31 (40.3) 34 (39.1) 20 (48.8) 44.7% 44.2% 41.5% 34.8%
Treatment line 0.057 0.03 0.71 0.55
   1 4 (11.1) 17 (22.1) 20 (23.0) 6 (14.6) 14.4% 18.5% 19.7% 15.2%
   2 20 (55.6) 25 (32.5) 43 (49.4) 14 (34.1) 41.6% 40.2% 40.3% 39.2%
   ≥3 12 (33.4) 35 (45.4) 24 (27.6) 21 (51.2) 44% 41.3% 40.1% 45.5%
Treatment regimen 0.40 <0.001 0.81 0.48
   Monotherapy 27 (75.0) 63 (81.8) 79 (90.8) 26 (63.4) 77.4% 78.7% 79.6% 83%
   Combined therapy 9 (25.0) 14 (18.2) 8 (9.2) 15 (36.6) 22.6% 21.3% 20.4% 17%

Data are presented as n (%), mean ± SD, or percentage. IPW, inverse probability weighting; pNEN, pancreatic neuroendocrine neoplasm; epNEN, extrapancreatic neuroendocrine neoplasm; Suru, surufatinib; Suni, sunitinib; Eve, everolimus; SD, standard deviation; ECOG-PS, Eastern Cooperative Oncology Group performance status; SSA, somatostatin analogues; anti-ang, anti-angiogenesis; mTORi, mTOR inhibitor.

Clinical treatment

In the IPW-adjusted cohort, nearly 80% of pNEN and epNEN patients received monotherapy in the two treatment groups, and the remaining patients were treated with combination therapy including SSAs, targeted agents, chemotherapy, and immune checkpoint inhibitors (ICIs).

Figure 2 shows the initial and final dose of the three targeted agents at IPW-adjusted cohort. Less patients received the standard dose initially and throughout the whole treatment course in the surufatinib group with 300 mg/day compared to that in sunitinib group with 37.5 mg/day and everolimus group with 10 mg/day (P<0.001). Through the treatment course, in the pNEN population, surufatinib’s dose of 250 mg increased from 22.4% to 49.5%, whereas that of 200 mg decreased from 66.3% to 45.1%. Sunitinib’s 25 mg dose rose from 19.5% to 34.8%, and that of 37.5 mg fell from 80.5% to 63.8%. Everolimus saw a rise in the 10 mg dose from 35.3% to 59.0%, and a drop in 5 mg from 59.3% to 16.4%. Through the treatment course, surufatinib’s 250 mg dose increased from 2.5% to 9%, and 200 mg decreased from 86.3% to 76.2% in epNEN patients. Everolimus’s 10 mg dose increased from 29.5% to 54.5%, and that of 5 mg decreased from 52.7% to 18.8%.

Figure 2 Initial and final dose of targeted agent during treatment course in (A) pNEN and (B) epNEN patients at IPW-adjusted cohort. pNEN, pancreatic neuroendocrine neoplasm; epNEN, extrapancreatic neuroendocrine neoplasm; IPW, inverse probability weighting.

Efficacy

In the unadjusted and IPW-adjusted cohort, surufatinib showed significantly longer median PFS (mPFS) than did sunitinib + everolimus in pNEN patients with the median follow-up time as 11 and 15 months, respectively (unadjusted cohort: 8.67 vs. 6.37 months, HR 0.587, 95% CI: 0.372–0.927, P=0.02; IPW-adjusted cohort: 8.30 vs. 6.33 months, HR 0.592, 95% CI: 0.436–0.804, P<0.001) (Figure 3A,3B). For epNEN patients, surufatinib showed significantly superior mPFS benefit over everolimus after IPW adjustment with the median follow-up time as 10 and 13 months, respectively (unadjusted cohort: 9.47 vs. 6.70 months, HR 0.728, 95% CI: 0.471–1.126, P=0.15; IPW-adjusted cohort: 8.73 vs. 3.70 months, HR 0.608, 95% CI: 0.456–0.810, P<0.001) (Figure 3C,3D). In addition, ORR and DCR were also found to have significant differences between targeted agent groups in pNEN and epNEN patients (Table 2).

Figure 3 Kaplan-Meier curves for progression-free survival in (A) pNEN patients at unadjusted cohort, (B) pNEN patients at IPW-adjusted cohort, (C) epNEN patients at unadjusted cohort, (D) epNEN patients at IPW-adjusted cohort. CI, confidence interval; pNEN, pancreatic neuroendocrine neoplasm; IPW, inverse probability weighting; epNEN, extrapancreatic neuroendocrine neoplasm.

Table 2

Treatment response with targeted agents in IPW-adjusted cohort

Treatment response pNEN (%) epNEN (%)
Suru Suni + Eve P value Suru Eve P value
Best response <0.001
   PR 21.0 9.9 0.004 6.2 5.6
   SD 70.2 65.7 79.7 58.4
   PD 8.8 24.3 14.1 36.0
ORR 21.0 9.9 0.03 6.2 5.6 0.84
DCR 91.2 75.7 0.004 85.9 64.0 <0.001

IPW, inverse probability weighting; pNEN, pancreatic neuroendocrine neoplasm; epNEN, extrapancreatic neuroendocrine neoplasm; Suru, surufatinib; Suni, sunitinib; Eve, everolimus; PR, partial response; SD, stable disease; PD, progressive disease; ORR, objective response rate; DCR, disease control rate.

Subgroup analysis of PFS in the IPW-adjusted cohort is shown in Figure 4 and demonstrated surufatinib’s notable survival benefits over controls. In pNEN, mPFS was 11.3 months with Ki67 3–20%, to 11.87 months for first-line, 12.63 months for initial full dose, and 13.13 months with dose adjustment. In epNEN, Ki67 3–20% had a mPFS of 10.4 months, first-line was 12.63 months, initial full dose was 9.47 months, dose adjustment was 11.1 months, and lung/thymus was 10.4 months.

Figure 4 Forest plot of subgroup analysis for progression-free survival in (A) pNEN and (B) epNEN patients at IPW-adjusted cohort. PFS, progression-free survival; CI, confidence interval; ECOG-PS, Eastern Cooperative Oncology Group performance status; TNM, tumor, node, metastasis; SSA, somatostatin analogues; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; pNEN, pancreatic neuroendocrine neoplasm; epNEN, extrapancreatic neuroendocrine neoplasm; IPW, inverse probability weighting.

To define whether certain clinical features impacted on PFS in NEN patients with targeted agents, univariate and multivariate analysis were performed (Tables 3,4). Multivariate Cox analysis revealed that male, functional status, Ki67 index >20%, previous SSA and germline mutation, poor differentiation at histopathology, liver metastasis, and multi treatment lines had a negative effect on PFS in pNEN and epNEN patients, respectively, whereas previous mTORi, dose adjustment, and surufatinib correlated with longer PFS.

Table 3

Univariate and multivariate analysis on clinical features associated with PFS in pNEN patients at IPW-adjusted cohort

Clinical features (surufatinib vs. sunitinib + everolimus) HR (95% CI) P value
Univariate analysis
   Male vs. female 1.33 (0.98−1.80) 0.06
   Age ≥65 vs. <65 years 1.02 (0.70−1.49) 0.90
   ECOG-PS ≥2 vs. 0–1 NR (0.00−NR) >0.99
   Functional vs. non-functional 1.85 (1.07−3.19) 0.02
   Germline mutated vs. sporadic 0.92 (0.23−3.74) 0.91
   Ki67 index
    3–20% vs. <3% 2.07 (0.40−10.77) 0.38
    >20% vs. <3% 6.20 (1.16−33.24) 0.03
   Distant metastatic vs. locally advanced 0.57 (0.25−1.31) 0.18
   Metastatic site number ≥3 vs. 0–2 1.68 (1.07−2.64) 0.02
   Liver metastasis (yes vs. no) 0.87 (0.44−1.73) 0.69
   Bone metastasis (yes vs. no) 0.80 (0.53−1.23) 0.31
   Treatment line
    Second line vs. first line 1.17 (0.74−1.84) 0.50
    ≥3rd line vs. first line 1.32 (0.84−2.09) 0.22
   Prior surgery (yes vs. no) 1.19 (0.88−1.60) 0.27
   Previous SSA (yes vs. no) 1.35 (0.98−1.85) 0.06
   Previous chemotherapy (yes vs. no) 1.26 (0.93−1.72) 0.13
   Previous anti-angiogenesis agents (yes vs. no) 0.85 (0.59−1.22) 0.37
   Previous mTORi (yes vs. no) 0.39 (0.19−0.81) 0.01
   Surufatinib vs. sunitinib + everolimus 0.59 (0.44−0.80) <0.001
   Combination therapy vs. monotherapy 0.75 (0.52−1.09) 0.12
   Initial standard dose (yes vs. no) 1.09 (0.80−1.47) 0.58
   Dose adjustment (yes vs. no) 0.50 (0.35−0.70) <0.001
   Dose interruption (yes vs. no) 0.84 (0.60−1.19) 0.33
Multivariate analysis
   Male vs. female 1.75 (1.25−2.44) 0.001
   Functional vs. non-functional 2.09 (1.16−3.74) 0.01
   Ki67 index >20% vs. <3% 12.74 (2.26−71.95) 0.004
   Surufatinib vs. sunitinib + everolimus 0.58 (0.42−0.79) <0.001
   Previous mTORi (yes vs. no) 0.48 (0.23−0.99) 0.04
   Previous SSA (yes vs. no) 1.73 (1.24−2.40) 0.001
   Dose adjustment (yes vs. no) 0.63 (0.45−0.89) 0.009

PFS, progression-free survival; pNEN, pancreatic neuroendocrine neoplasm; IPW, inverse probability weighting; HR, hazard ratio; CI, confidence interval; ECOG-PS, Eastern Cooperative Oncology Group performance status; NR, not reported; SSA, somatostatin analogues; mTORi, mTOR inhibitor.

Table 4

Univariate and multivariate analysis on clinical features associated with PFS in epNEN patients at IPW-adjusted cohort

Clinical features (surufatinib vs. sunitinib + everolimus) HR (95% CI) P value
Univariate analysis
   Male vs. female 1.00 (0.75−1.33) 0.99
   Age ≥65 vs. <65 years 1.03 (0.72−1.47) 0.86
   ECOG-PS ≥2 vs. 0–1 1.11 (0.51−2.42) 0.79
   Primary tumor site
    Lung/thymus/mediastinum vs. GI 1.18 (0.78−1.79) 0.43
    Others vs. GI 0.91 (0.65−1.27) 0.58
   Functional vs. non-functional 0.55 (0.24−1.29) 0.17
   Germline mutated vs. sporadic 2.62 (1.74−3.95) <0.001
   Poorly-differentiated vs. well-differentiated 5.04 (3.14−8.08) <0.001
   Ki67 index
    3–20% vs. <3% 1.55 (0.78−3.09) 0.20
    >20% vs. <3% 3.42 (1.66−7.03) <0.001
   Distant metastatic vs. locally advanced 0.77 (0.48−1.25) 0.28
   Metastatic site number ≥3 vs. 0–2 1.09 (0.78−1.52) 0.61
   Liver metastasis (yes vs. no) 1.34 (1.00−1.79) 0.04
   Bone metastasis (yes vs. no) 0.76 (0.57−1.02) 0.06
   Treatment line
    Second line vs. first line 2.08 (1.29−3.35) 0.003
    ≥3rd line vs. first line 2.41 (1.51−3.84) <0.001
   Prior surgery (yes vs. no) 0.92 (0.69−1.22) 0.56
   Previous SSA (yes vs. no) 1.02 (0.76−1.36) 0.90
   Previous chemotherapy (yes vs. no) 1.68 (1.24−2.29) <0.001
   Previous anti-angiogenesis agents (yes vs. no) 1.08 (0.77−1.51) 0.67
   Previous mTORi (yes vs. no) 0.77 (0.46−1.29) 0.31
   Surufatinib vs. everolimus 0.61 (0.46−0.81) <0.001
   Combination therapy vs. monotherapy 1.24 (0.89−1.74) 0.21
   Initial standard dose (yes vs. no) 1.38 (0.99−1.94) 0.058
   Dose adjustment (yes vs. no) 0.78 (0.57−1.08) 0.13
   Dose interruption (no vs. yes) 1.63 (1.11−2.39) 0.01
Multivariate analysis
   Germline mutated vs. sporadic 5.62 (3.49−9.04) <0.001
   Poorly-differentiated vs. well-differentiated 7.45 (4.40−12.61) <0.001
   Liver metastasis (yes vs. no) 1.72 (1.24−2.39) 0.001
   Treatment line
    Second line vs. first line 1.62 (1.00−2.62) 0.04
    ≥3rd line vs. first line 1.88 (1.16−3.06) 0.01
   Surufatinib vs. everolimus 0.62 (0.45−0.84) 0.002

PFS, progression-free survival; epNEN, extrapancreatic neuroendocrine neoplasm; IPW, inverse probability weighting; HR, hazard ratio; CI, confidence interval; ECOG-PS, Eastern Cooperative Oncology Group performance status; GI, gastro-intestinal neuroendocrine neoplasms; SSA, somatostatin analogues; mTORi, mTOR inhibitor.

Safety

Overall, no significant differences were found in the frequency of treatment-related AE with surufatinib, sunitinib, and everolimus in pNEN and epNEN patients (P=0.46 and 0.42) (Tables 5,6). In the pNEN cohort, the reported incidence of grade 3 or higher treatment-related AE were 26.0% for sunitinib, 25% for surufatinib, and 11.1% for everolimus (Table 5). Within the epNEN group, such AEs occurred in 31% of patients treated with surufatinib and 17.1% with everolimus (Table 6). The spectrum of AEs varied between targeted agents. The most frequent AEs were proteinuria, hypertension, increased blood thyroid-stimulating hormone, increased bilirubin, and hyperuricemia with surufatinib; hematologic toxicity (decreased platelet count, deceased white blood cell, decreased neutrophil count, and anemia) with sunitinib; anemia, hyperglycemia, hyperlipemia, stomatitis, and increased liver enzymes (γ-glutamyl transferase, aspartate aminotransferase) with everolimus. Moreover, the most common grade 3 or >3 AE that patients experienced was hypertension with surufatinib, which was typically controlled with medical management; decreased neutrophil count with sunitinib and anemia with everolimus were also effectively managed as per clinical guidelines. Apart from that, diarrhea (P=0.03), hypothyroidism (P=0.02), and occult blood positive (P=0.02) were found more commonly with surufatinib, whereas interstitial pneumonia (P=0.009) was more likely to appear with everolimus.

Table 5

Adverse events with targeted agents in pNEN patients at unadjusted cohort

Adverse events Surufatinib, n=36, n (%) Sunitinib, n=50, n (%) Everolimus, n=27, n (%) P value (any grade) P value (grade ≥3)
Any grade Grade ≥3 Any grade Grade ≥3 Any grade Grade ≥3
Any AE 31 (86.1) 9 (25.0) 39 (78.0) 13 (26.0) 20 (74.1) 3 (11.1) 0.46 0.28
Hypertension 6 (16.7) 1 (2.8) 9 (18.0) 3 (6.0) 0 0 0.03 0.56
Proteinuria 8 (22.2) 0 6 (12.0) 0 0 0 0.01 NA
WBC decreased 2 (5.6) 0 17 (34.0) 0 2 (7.4) 0 <0.001 NA
Neutrophil decreased 1 (2.8) 0 15 (30.0) 6 (12.0) 1 (3.7) 0 <0.001 0.01
Febrile neutropenia 0 0 1 (2.0) 1 (2.0) 0 0 >0.99 >0.99
LYM decreased 5 (13.9) 0 2 (4.0) 0 4 (14.8) 0 0.16 NA
Anemia 4 (11.1) 2 (5.6) 11 (22.0) 0 8 (29.6) 0 0.18 0.15
PLT decreased 4 (11.1) 0 19 (38.0) 1 (2.0) 2 (7.4) 0 0.001 >0.99
Hemorrhage 2 (5.6) 1 (2.8) 2 (4.0) 1 (2.0) 1 (3.7) 0 >0.99 >0.99
Occult blood positive 5 (13.9) 0 1 (2.0) 0 0 0 0.02 NA
AST increased 4 (11.1) 2 (5.6) 4 (8.0) 0 7 (25.9) 1 (3.7) 0.09 0.17
ALT increased 3 (8.3) 1 (2.8) 6 (12.0) 0 7 (25.9) 1 (3.7) 0.11 0.30
ALP increased 5 (13.9) 0 2 (4.0) 0 3 (11.1) 0 0.26 NA
GGT increased 3 (8.3) 0 1 (2.0) 0 7 (25.9) 2 (7.4) 0.003 0.055
LDH increased 4 (11.1) 0 3 (6.0) 0 4 (14.8) 0 0.36 NA
Bilirubin increased 11 (30.6) 2 (5.6) 3 (6.0) 0 2 (7.4) 0 0.003 0.15
Hypoalbuminemia 4 (11.1) 0 6 (12.0) 0 5 (18.5) 0 0.72 NA
Edema 0 0 5 (10.0) 0 1 (3.7) 0 0.08 NA
Creatinine increased 3 (8.3) 0 4 (8.0) 0 1 (3.7) 0 0.80 NA
Hyperuricemia 6 (16.7) 0 1 (2.0) 0 1 (3.7) 0 0.02 NA
Hyperlipemia 4 (11.1) 1 (2.8) 0 0 7 (25.9) 0 <0.001 0.55
Hyperglycemia 7 (19.4) 0 0 0 8 (29.6) 0 <0.001 NA
Hypoglycemia 1 (2.8) 0 1 (2.0) 0 0 0 >0.99 NA
TSH increased 6 (16.7) 0 1 (2.0) 0 0 0 0.006 NA
Hypothyroidism 3 (8.3) 0 0 0 0 0 0.03 NA
Nausea/vomiting 2 (5.6) 0 0 0 1 (3.7) 0 0.17 NA
Diarrhea 6 (16.7) 0 1 (2.0) 0 1 (3.7) 0 0.02 NA
Abdominal discomfort 1 (2.8) 0 1 (2.0) 0 0 0 >0.99 NA
Stomatitis 0 0 3 (6.0) 0 7 (25.9) 0 <0.001 NA
Muscle pain 0 0 0 0 0 0 NA NA
Fatigue 0 0 2 (4.0) 0 0 0 0.50 NA
HFS reaction 1 (2.8) 0 5 (10.0) 2 (4.0) 0 0 0.16 0.50
Cough 0 0 0 0 1 (3.7) 0 0.23 NA
Interstitial pneumonia 0 0 0 0 1 (3.7) 0 0.23 NA
Infection 1 (2.8) 0 1 (2.0) 1 (2.0) 3 (11.1) 1 (3.7) 0.15 0.71
Cardiac toxicity 1 (2.8) 0 0 0 1 (3.7) 0 0.30 NA

AE, adverse event; pNEN, pancreatic neuroendocrine neoplasm; WBC, white blood cell; LYM, lymphocyte; PLT, platelet; AST, aspartate aminotransferase; ALT, alanine aminotransferase; ALP, alkaline phosphatase; GGT, γ-glutamyl transferase; LDH, lactate dehydrogenase; TSH, thyroid-stimulating hormone; HFS, hand-foot skin syndrome; NA, not available.

Table 6

Adverse events with targeted agents in epNEN patients at unadjusted cohort

Adverse events Surufatinib, n=87, n (%) Everolimus, n=41, n (%) P value (any grade) P value (grade ≥3)
Any grade Grade ≥3 Any grade Grade ≥3
Any AE 73 (83.9) 27 (31.0) 32 (78.0) 7 (17.1) 0.42 0.09
Hypertension 30 (34.5) 14 (16.1) 0 0 <0.001 0.005
Proteinuria 33 (37.9) 2 (2.3) 1 (2.4) 0 <0.001 >0.99
WBC decreased 7 (8.0) 2 (2.3) 4 (9.8) 0 0.74 >0.99
Neutrophil decreased 3 (3.4) 1 (1.1) 3 (7.3) 2 (4.9) 0.38 0.24
Febrile neutropenia 0 0 1 (2.4) 1 (2.4) 0.32 0.32
LYM decreased 12 (13.8) 2 (2.3) 7 (17.1) 0 0.62 >0.99
Anemia 15 (17.2) 3 (3.4) 15 (36.6) 3 (7.3) 0.01 0.38
PLT decreased 6 (6.9) 3 (3.4) 4 (9.8) 0 0.72 0.55
Hemorrhage 6 (6.9) 1 (1.1) 1 (2.4) 0 0.42 >0.99
Occult blood positive 5 (5.7) 0 0 0 0.17 NA
AST increased 16 (18.4) 2 (2.3) 7 (17.1) 1 (2.4) 0.85 >0.99
ALT increased 9 (10.3) 2 (2.3) 4 (9.8) 0 >0.99 >0.99
ALP increased 4 (4.6) 1 (1.1) 5 (12.2) 0 0.14 >0.99
GGT increased 8 (9.2) 3 (3.4) 7 (17.1) 1 (2.4) 0.24 >0.99
LDH increased 18 (20.7) 0 9 (22.0) 0 0.87 NA
Bilirubin increased 17 (19.5) 3 (3.4) 1 (2.4) 0 0.009 0.55
Hypoalbuminemia 12 (13.8) 0 5 (12.2) 0 0.80 NA
Edema 1 (1.1) 0 0 0 >0.99 NA
Creatinine increased 9 (10.3) 0 2 (4.9) 1 (2.4) 0.50 0.32
Hyperuricemia 18 (20.7) 1 (1.1) 1 (2.4) 0 0.007 >0.99
Hyperlipemia 7 (8.0) 0 10 (24.4) 0 0.01 NA
Hyperglycemia 8 (9.2) 0 15 (36.6) 0 <0.001 NA
Hypoglycemia 3 (3.4) 0 0 0 0.55 NA
TSH increased 22 (25.3) 0 0 0 <0.001 NA
Hypothyroidism 3 (3.4) 0 0 0 0.55 NA
Nausea/vomiting 2 (2.3) 0 1 (2.4) 0 >0.99 NA
Diarrhea 13 (14.9) 0 2 (4.9) 0 0.14 NA
Abdominal discomfort 1 (1.1) 0 0 0 >0.99 NA
Stomatitis 0 0 9 (22.0) 1 (2.4) <0.001 0.32
Muscle pain 1 (1.1) 0 0 0 >0.99 NA
Fatigue 6 (6.9) 0 2 (4.9) 0 >0.99 NA
HFS reaction 1 (1.1) 0 2 (4.9) 0 0.24 NA
Cough 0 0 1 (2.4) 0 0.32 NA
Interstitial pneumonia 0 0 4 (9.8) 0 0.009 NA
Infection 1 (1.1) 0 2 (4.9) 0 0.24 NA
Cardiac toxicity 1 (1.1) 0 2 (4.9) 0 0.24 NA

AE, adverse event; epNEN, extrapancreatic neuroendocrine neoplasm; WBC, white blood cell; LYM, lymphocyte; PLT, platelet; AST, aspartate aminotransferase; ALT, alanine aminotransferase; ALP, alkaline phosphatase; GGT, γ-glutamyl transferase; LDH, lactate dehydrogenase; TSH, thyroid-stimulating hormone; HFS, hand-foot skin syndrome; NA, not available.


Discussion

NEN is a neoplasm with extremely high heterogeneity, which in some cases can be mixed with non-neuroendocrine components, resulting in significant differences in biological behavior, malignant potential and prognosis of NEN (25). NENs have been found in multiple organs, of which GEP-NENs are more common and thymic/lung NEN appear relatively rare (6,26). The development and advancement of targeted therapies have revolutionized the treatment landscape for advanced NENs, with endorsement by worldwide national treatment guidelines. To our knowledge, this is the first study to compare the efficacy and safety of surufatinib, sunitinib, and everolimus, all of which have been approved as standard treatment for advanced NENs patients in China. With high heterogeneity of NEN, it is critical to explore whether various clinical subtypes could benefit from current targeted agents in the real world.

Our study demonstrated that surufatinib (pNEN: 8.30 months, epNEN: 8.73 months) showed significantly superior mPFS benefit over sunitinib and everolimus in the overall population. Data on surufatinib, a targeted agent originally researched and launched in China, warrant further explorations currently. In the SANET-p and SANET-ep trials, the mPFS was 10.9 months (95% CI: 7.5–13.8) and 9.2 months (95% CI: 7.4–11.1), respectively, for surufatinib, which were slightly longer than that in our study (16,17). Our approach, unlike clinical trials, did not impose strict entry criteria to better mirror real-world scenarios, accommodating patients with ECOG-PS ≥2, primarily ≥2 lines of treatment, Ki67% ≥20%, advanced pathological grades, poor differentiation, and uncommon/multiple primary tumor sites, potentially contributing to the shorter mPFS than that in previous studies comparing sunitinib and everolimus (27,28). In addition, we did not exclude patients receiving combination therapy with other agents, including SSAs, chemotherapy, and ICIs to meet practical treatment demands, although survival periods showed no statistical differences compared with monotherapy. Combination therapy has become a common medication pattern in the current tumor treatment area. Several RCTs had been developed to explore whether everolimus plus SSA could improve effectiveness in advanced NENs, but the desired results were not achieved with no adequate improvement on PFS and overall survival (OS). The RADIANT-2 trial demonstrated that everolimus plus octreotide long-acting repeatable (LAR) obtained mPFS (16.4 vs. 11.3 months, HR 0.77, 95% CI: 0.59–1.00, one-sided P=0.02) and OS (29.2 vs. 35.2 months, HR 1.17, 95% CI: 0.92–1.49) with no statistical significances in advanced NENs and carcinoid syndrome patients (29,30), whereas just in the colorectal NENs subgroup this combined treatment was associated with a 66% reduction in the estimated risk for disease progression (29.90 vs. 6.57 months, HR 0.34, 95% CI: 0.13–0.89, P=0.01) (31). The COOPERATE-2 trial showed that the addition of pasireotide to everolimus was not associated with the improvement in PFS compared with everolimus alone (16.8 vs. 16.6 months, HR 0.99, 95% CI: 0.64–1.54, P=0.48) (32). For sunitinib and surufatinib, evidences on combination therapy were limited. A single-arm phase I trial evaluated that the overall ORR and DCR were 23.8% and 76.2% in 21 NEN patients (more than half were NECs) receiving surufatinib combined with toripalimab (a programmed cell death protein-1inhibitor) (33), which provided a potential option for higher malignancy subtypes. With regard to targeted agents, further studies are warranted to confirm whether combination therapy can bring more benefits in advanced NENs.

In this study, some histopathological factors associated with tumor grading/classification/staging (differentiation, Ki67 index, liver metastasis, functional status, germline mutation) were identified as related with mPFS in advanced NEN patients, which had been also validated by numerous studies (34-37). Phase III trials of targeted therapy solely encompassed patients with G1/G2 NENs, while the effectiveness and safety of drugs for high-grade (G3 or Ki67 >20%) NENs and poorly-differentiation NECs remain unexplored. Two prospective phase 2 trials tried to verify everolimus and sunitinib activity in advanced G3/NEC patients and found that efficacy was limited with mPFS of 1.2 and 1.4 months (38,39), respectively, whereas surufatinib achieved a 23% ORR and 4.14 months mPFS in NEC (40). Hence, application of targeted agents needs careful consideration. Liver metastasis was considered a common influencing factor (5,41), while compared to the presence of liver metastasis itself, the liver tumor burden may serve as a more instructive prognostic factor (42,43). A retrospective study found that liver tumor burden ≤25% (HR 0.27, 95% CI: 0.13–0.55, P<0.001) became the significant factors influencing OS in pNENs treated with everolimus (44), which could offer limited proof on availability of therapeutic effect prediction. Apart from that, we also found that functional and germline-mutated NENs, subgroups that clinical trials commonly excluded, were related with worse PFS. A small portion of NENs (approximately 10%) were related to genetic factors, namely those carrying germline gene mutations and occurring hereditary syndromes (45). Nuñez et al. found that everolimus achieved similar efficacy in multiple endocrine neoplasia type 1 (MEN1) and Von-Hippel-Lindau (VHL) compared with sporadic NENs (46). This differed from the conclusion we drew, but considering the small population, more evidences would be needed. Regarding treatment-related factors, previous SSA was considered to increase the risk of disease progression, whereas RADIANT-4 subgroup analysis indicated that everolimus had not significantly improved PFS in any previous SSA subgroup (47). NCCN, European Society for Medical Oncology (ESMO), and ENETS guidelines recommend SSAs as the first-line treatment of patients with somatostatin receptor (SSTR) positivity, G1/2 NENs with relatively low proliferative activity (Ki67 <10%), low tumor burden, and indolent behavior (11,12,48). In the absence of high-level evidences, systemic therapy selection and sequencing in advanced NENs are not fully elucidated, which mainly depends on the biological behavior and malignancy of the tumor, patient characteristics and treatment goals (49). Multiple prior treatment lines tended to be unfavorable factors on PFS in our study, in similarity with conclusions drawn in other investigations (27,28,50), which was probably due to the powerful invasiveness of an uncontrollable tumor.

A considerable portion of patients received surufatinib, sunitinib, and everolimus at reduced doses initially in this study. Although combination therapy possibly induced physicians to further take tolerance into account, more than 60% of patients with monotherapy did not accept standard dose initially. We analyzed that the initial recommended dose for treatment would not affect PFS and dose adjustment may play a positive role on PFS in pNEN patients. It is common that the strategy with lower tolerant initial dose and gradually increased dose is chosen to avoid AEs as far as possible in clinical practice. For small molecule kinase inhibitors, dose escalation is indeed one of the methods to optimize administration strategy. The DESIREE prospective trial reported that the use of a stepwise dose-escalation schedule of everolimus (2.5 mg week 1 increased by 2.5 mg per week to 10 mg) could significantly reduce the incidence of high-grade stomatitis in patients with hormone receptor (HR)+/human epidermal growth factor receptor 2 (HER2) metastatic breast cancer (51). Escalation of sunitinib dosing (12.5 mg steps) in patients with metastatic renal cell cancer, based on tolerable toxicity, has been shown to be safe and improve outcome (52). Despite lacking related evidences in advanced NENs, dose escalation of targeted agents can be a reasonable attempt in real-world practice. As a result, dose adjustment appeared in nearly one-third of patients in an effort to balance efficacy and tolerance. Berardi et al. reported that cumulative dose >3,000 mg and dose intensity >9 mg/day of everolimus played a prognostic role for patients with advanced pancreatic neuroendocrine tumors (pNETs) (53). Despite that a small sample-size study suggested that median daily dose reduction of sunitinib would probably not alter prognosis and safety for patients with advanced pNENs (54), Lee et al. demonstrated that severe AEs occurred in 63% of patients with sunitinib whose prolonged treatment period was significantly correlated with decreased relative dose intensity (55). Consequently, well-developed patient management with appropriate initial dose selection, dose adjustment, and interruption will provide guarantees for the long-term application of targeted agents.

In general, the safety profile of surufatinib, sunitinib, and everolimus was substantially similar to the reports of RCTs (16-20), which illustrated that under appropriate management, combination therapy would not increase the potential toxicity. Except adverse events of special interest (AESI) for the three drugs, there were differences in common AEs including hematologic toxicity and hepatotoxicity. Hematologic toxicity, which mainly manifested as myelocyte and megakaryocyte suppression, occurred more frequently with sunitinib, whereas everolimus and surufatinib were more likely to cause erythrocytes suppression and hemorrhage, respectively. Hepatotoxicity was also represented as different abnormal liver function indexes among the drugs. Considering that there is currently no clear evidence for position and sequence among targeted agents, it is recommended to match baseline characteristics of patients with drug toxicity for selection in clinical settings.

Our study had some limitations. Firstly, the most important constraint was the retrospective observational design with inherent bias, although we applied statistical methods to balance heterogeneity as far as possible. The relatively small sample size probably expanded the negative impact, so that we collected substantial variables and conducted subgroup analysis in order to explore the outcomes under different clinical features. Moreover, clinical data capture based on medical records system, together with early approval of sunitinib and everolimus in China, may have resulted in patient loss of follow-up and biased survival outcome. What’s more, surufatinib’s approval period was much shorter than everolimus and sunitinib, which resulted in differences on patients’ follow-up time and survival outcome acquiring. Under these circumstances, prospective research is required for further verification and OS data would be reported. Finally, we included NENs originating from any organ and tissue to reflect a real-world setting, yet the intrinsic heterogeneity possibly confused the results considering drug indication and histopathological difference.


Conclusions

This study explored the efficacy and safety of surufatinib, sunitinib, and everolimus in advanced NEN patients in real-world clinical practice in China. We demonstrated that surufatinib significantly prolonged PFS compared to sunitinib and everolimus in patients with advanced NEN treatments with different AE types to provide future perspective in patient management. This finding, together with identified clinical features affecting survival outcome, may lead to implications for selection of targeted agents in Chinese advanced NEN patients. These results should be further validated in well-designed prospective studies, in which specific molecular biomarkers should be explored to facilitate personalized treatment progress for advanced NEN.


Acknowledgments

Funding: This study was supported by the 2022 Shanghai Youth Pharmacy Talent Ability Enhancement Project “Intelligent clinical comprehensive evaluation platform for anti-tumor drugs” (to Q.D.), and the 2022–2023 Shanghai Anticancer Association “Soaring Project” (No. SHAX-QT-202101 to Q.Z.).


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://jgo.amegroups.com/article/view/10.21037/jgo-24-218/rc

Data Sharing Statement: Available at https://jgo.amegroups.com/article/view/10.21037/jgo-24-218/dss

Peer Review File: Available at https://jgo.amegroups.com/article/view/10.21037/jgo-24-218/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jgo.amegroups.com/article/view/10.21037/jgo-24-218/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). This study was approved by institutional ethics committee of Fudan University Shanghai Cancer Center (No. 2205254-19) and individual consent for this retrospective analysis was waived.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Ma X, Ding Y, Li W, et al. Diagnosis and management of gastroenteropancreatic neuroendocrine neoplasms by nuclear medicine: Update and future perspective. Front Oncol 2022;12:1061065. [Crossref] [PubMed]
  2. Taal BG, Visser O. Epidemiology of neuroendocrine tumours. Neuroendocrinology 2004;80:3-7. [Crossref] [PubMed]
  3. Lee MR, Harris C, Baeg KJ, et al. Incidence Trends of Gastroenteropancreatic Neuroendocrine Tumors in the United States. Clin Gastroenterol Hepatol 2019;17:2212-2217.e1. [Crossref] [PubMed]
  4. Fan JH, Zhang YQ, Shi SS, et al. A nation-wide retrospective epidemiological study of gastroenteropancreatic neuroendocrine neoplasms in china. Oncotarget 2017;8:71699-708. [Crossref] [PubMed]
  5. Dasari A, Shen C, Halperin D, et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol 2017;3:1335-42. [Crossref] [PubMed]
  6. Hofland J, Kaltsas G, de Herder WW. Advances in the Diagnosis and Management of Well-Differentiated Neuroendocrine Neoplasms. Endocr Rev 2020;41:371-403. [Crossref] [PubMed]
  7. Cai W, Tan Y, Ge W, et al. Pattern and risk factors for distant metastases in gastrointestinal neuroendocrine neoplasms: a population-based study. Cancer Med 2018;7:2699-709. [Crossref] [PubMed]
  8. Riihimäki M, Hemminki A, Sundquist K, et al. The epidemiology of metastases in neuroendocrine tumors. Int J Cancer 2016;139:2679-86. [Crossref] [PubMed]
  9. La Salvia A, Espinosa-Olarte P, Riesco-Martinez MDC, et al. Targeted Cancer Therapy: What's New in the Field of Neuroendocrine Neoplasms? Cancers (Basel) 2021;13:1701. [Crossref] [PubMed]
  10. Wu WM, Chen J, Bai CM, et al. The Chinese guidelines for the diagnosis and treatment of pancreatic neuroendocrine neoplasms (2020). Zhonghua Wai Ke Za Zhi 2021;59:401-21. [Crossref] [PubMed]
  11. Shah MH, Goldner WS, Benson AB, et al. Neuroendocrine and Adrenal Tumors, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021;19:839-68. [Crossref] [PubMed]
  12. Zandee WT, de Herder WW. The Evolution of Neuroendocrine Tumor Treatment Reflected by ENETS Guidelines. Neuroendocrinology 2018;106:357-65. [Crossref] [PubMed]
  13. Carrasco P, Zuazo-Gaztelu I, Casanovas O. Sprouting strategies and dead ends in anti-angiogenic targeting of NETs. J Mol Endocrinol 2017;59:R77-91. [Crossref] [PubMed]
  14. Lauricella E, Mandriani B, Cavallo F, et al. Angiogenesis in NENs, with a focus on gastroenteropancreatic NENs: from biology to current and future therapeutic implications. Front Oncol 2022;12:957068. [Crossref] [PubMed]
  15. Scoazec JY. Angiogenesis in neuroendocrine tumors: therapeutic applications. Neuroendocrinology 2013;97:45-56. [Crossref] [PubMed]
  16. Xu J, Shen L, Bai C, et al. Surufatinib in advanced pancreatic neuroendocrine tumours (SANET-p): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2020;21:1489-99. [Crossref] [PubMed]
  17. Xu J, Shen L, Zhou Z, et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2020;21:1500-12. [Crossref] [PubMed]
  18. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011;364:501-13. [Crossref] [PubMed]
  19. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 2011;364:514-23. [Crossref] [PubMed]
  20. Yao JC, Fazio N, Singh S, et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet 2016;387:968-77. [Crossref] [PubMed]
  21. Ohmoto A, Morizane C. Genomic Profiles and Current Therapeutic Agents in Neuroendocrine Neoplasms. Curr Drug Targets 2020;21:389-405. [Crossref] [PubMed]
  22. Rindi G, Klimstra DS, Abedi-Ardekani B, et al. A common classification framework for neuroendocrine neoplasms: an International Agency for Research on Cancer (IARC) and World Health Organization (WHO) expert consensus proposal. Mod Pathol 2018;31:1770-86. [Crossref] [PubMed]
  23. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228-47. [Crossref] [PubMed]
  24. Freites-Martinez A, Santana N, Arias-Santiago S, et al. Using the Common Terminology Criteria for Adverse Events (CTCAE - Version 5.0) to Evaluate the Severity of Adverse Events of Anticancer Therapies. Actas Dermosifiliogr (Engl Ed) 2021;112:90-2. [Crossref] [PubMed]
  25. Wang F, Lou X, Qin Y, et al. Mixed neuroendocrine nonneuroendocrine neoplasms of the pancreas: a case report and literature review of pancreatic mixed neuroendocrine nonneuroendocrine neoplasm. Gland Surg 2021;10:3443-52. [Crossref] [PubMed]
  26. Lang M, Pausch TM, Anamaterou C. Thymic neuroendocrine neoplasms-what we know, what we don't know, and what to do about it. Transl Cancer Res 2023;12:1-3. [Crossref] [PubMed]
  27. Yoo C, Cho H, Song MJ, et al. Efficacy and safety of everolimus and sunitinib in patients with gastroenteropancreatic neuroendocrine tumor. Cancer Chemother Pharmacol 2017;79:139-46. [Crossref] [PubMed]
  28. Daskalakis K, Tsoli M, Angelousi A, et al. Anti-tumour activity of everolimus and sunitinib in neuroendocrine neoplasms. Endocr Connect 2019;8:641-53. [Crossref] [PubMed]
  29. Pavel ME, Baudin E, Öberg KE, et al. Efficacy of everolimus plus octreotide LAR in patients with advanced neuroendocrine tumor and carcinoid syndrome: final overall survival from the randomized, placebo-controlled phase 3 RADIANT-2 study. Ann Oncol 2017;28:1569-75. [Crossref] [PubMed]
  30. Pavel ME, Hainsworth JD, Baudin E, et al. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 2011;378:2005-12. [Crossref] [PubMed]
  31. Castellano D, Bajetta E, Panneerselvam A, et al. Everolimus plus octreotide long-acting repeatable in patients with colorectal neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-2 study. Oncologist 2013;18:46-53. [Crossref] [PubMed]
  32. Kulke MH, Ruszniewski P, Van Cutsem E, et al. A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial. Ann Oncol 2017;28:1309-15. [Crossref] [PubMed]
  33. Cao Y, Lu M, Sun Y, et al. Surufatinib plus toripalimab in patients with advanced solid tumors: a single-arm, open-label, phase 1 trial. J Cancer Res Clin Oncol 2023;149:779-89. [Crossref] [PubMed]
  34. Mizuno Y, Kudo A, Akashi T, et al. Sunitinib shrinks NET-G3 pancreatic neuroendocrine neoplasms. J Cancer Res Clin Oncol 2018;144:1155-63. [Crossref] [PubMed]
  35. Benslama N, Bollard J, Vercherat C, et al. Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors. Invest New Drugs 2016;34:654-62. [Crossref] [PubMed]
  36. Daddi N, Schiavon M, Filosso PL, et al. Prognostic factors in a multicentre study of 247 atypical pulmonary carcinoids. Eur J Cardiothorac Surg 2014;45:677-86. [Crossref] [PubMed]
  37. Chi Y, Du F, Zhao H, et al. Characteristics and long-term prognosis of patients with rectal neuroendocrine tumors. World J Gastroenterol 2014;20:16252-7. [Crossref] [PubMed]
  38. Okuyama H, Ikeda M, Okusaka T, et al. A Phase II Trial of Everolimus in Patients with Advanced Pancreatic Neuroendocrine Carcinoma Refractory or Intolerant to Platinum-Containing Chemotherapy (NECTOR Trial). Neuroendocrinology 2020;110:988-93. [Crossref] [PubMed]
  39. Pellat A, Dreyer C, Couffignal C, et al. Clinical and Biomarker Evaluations of Sunitinib in Patients with Grade 3 Digestive Neuroendocrine Neoplasms. Neuroendocrinology 2018;107:24-31. [Crossref] [PubMed]
  40. Lu M, Yu X, Chen Z, et al. 155P Surufatinib plus toripalimab for second-line treatment of advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma, esophageal squamous cell carcinoma (ESCC) and neuroendocrine carcinoma (NEC): A multicenter, single-arm phase II study. Ann Oncol 2021;32:S1448. [Crossref]
  41. Cao C, Yan TD, Kennedy C, et al. Bronchopulmonary carcinoid tumors: long-term outcomes after resection. Ann Thorac Surg 2011;91:339-43. [Crossref] [PubMed]
  42. Touloupas C, Faron M, Hadoux J, et al. Long Term Efficacy and Assessment of Tumor Response of Transarterial Chemoembolization in Neuroendocrine Liver Metastases: A 15-Year Monocentric Experience. Cancers (Basel) 2021;13:5366. [Crossref] [PubMed]
  43. Zener R, Yoon H, Ziv E, et al. Outcomes After Transarterial Embolization of Neuroendocrine Tumor Liver Metastases Using Spherical Particles of Different Sizes. Cardiovasc Intervent Radiol 2019;42:569-76. [Crossref] [PubMed]
  44. Kurita Y, Kobayashi N, Hara K, et al. Effectiveness and Prognostic Factors of Everolimus in Patients with Pancreatic Neuroendocrine Neoplasms. Intern Med 2023;62:159-67. [Crossref] [PubMed]
  45. Cives M, Strosberg J. An update on gastroenteropancreatic neuroendocrine tumors. Oncology (Williston Park) 2014;28:749-56, 758. [PubMed]
  46. Nuñez JE, Donadio M, Filho DR, et al. The efficacy of everolimus and sunitinib in patients with sporadic or germline mutated metastatic pancreatic neuroendocrine tumors. J Gastrointest Oncol 2019;10:645-51. [Crossref] [PubMed]
  47. Singh S, Carnaghi C, Buzzoni R, et al. Everolimus in Neuroendocrine Tumors of the Gastrointestinal Tract and Unknown Primary. Neuroendocrinology 2018;106:211-20. [Crossref] [PubMed]
  48. Pavel M, Öberg K, Falconi M, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020;31:844-60. [Crossref] [PubMed]
  49. Rodriguez-Freixinos V, Thawer A, Capdevila J, et al. Advanced Pancreatic Neuroendocrine Neoplasms: Which Systemic Treatment Should I Start With? Curr Oncol Rep 2021;23:80. [Crossref] [PubMed]
  50. Viúdez A, Crespo G, Gómez Dorronsoro ML, et al. Usefulness of an immunohistochemical score in advanced pancreatic neuroendocrine tumors treated with CAPTEM or everolimus. Pancreatology 2021;21:215-23. [Crossref] [PubMed]
  51. Schmidt M, Lübbe K, Decker T, et al. A multicentre, randomised, double-blind, phase II study to evaluate the tolerability of an induction dose escalation of everolimus in patients with metastatic breast cancer (DESIREE). ESMO Open 2022;7:100601. [Crossref] [PubMed]
  52. Maráz A, Cserháti A, Uhercsák G, et al. Dose escalation can maximize therapeutic potential of sunitinib in patients with metastatic renal cell carcinoma. BMC Cancer 2018;18:296. [Crossref] [PubMed]
  53. Berardi R, Torniai M, Pusceddu S, et al. Prognostic impact of the cumulative dose and dose intensity of everolimus in patients with pancreatic neuroendocrine tumors. Cancer Med 2017;6:1493-9. [Crossref] [PubMed]
  54. Matsui S, Kudo A, Ogura T, et al. Does sunitinib have a patient-specific dose without diminishing its antitumor effect on advanced pancreatic neuroendocrine neoplasms? J Cancer Res Clin Oncol 2019;145:2097-104. [Crossref] [PubMed]
  55. Lee L, Ito T, Igarashi H, et al. Dose and schedule modification are required for long-term continuation of sunitinib in Japanese patients with advanced pancreatic neuroendocrine tumors. Cancer Chemother Pharmacol 2018;81:163-9. [Crossref] [PubMed]
Cite this article as: Zhu L, Ye X, She Y, Liu W, Hasegawa K, Rossi RE, Du Q, Zhai Q. Assessing the effectiveness and safety of surufatinib versus everolimus or sunitinib in advanced neuroendocrine neoplasms: insights from a real-world, retrospective cohort study using propensity score and inverse probability treatment weighting analysis. J Gastrointest Oncol 2024;15(2):689-709. doi: 10.21037/jgo-24-218

Download Citation